1
|
Chen X, Huang M, Chen Y, Xu H, Wu M. Mineralocorticoid receptor antagonists and heart failure with preserved ejection fraction: current understanding and future prospects. Heart Fail Rev 2025; 30:191-208. [PMID: 39414721 DOI: 10.1007/s10741-024-10455-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2024] [Indexed: 10/18/2024]
Abstract
The mineralocorticoid receptor (MR), part of the steroid hormone receptor subfamily within nuclear hormone receptors, is found in the kidney and various non-epithelial tissues, including the heart and blood vessels. When improperly activated, it can contribute to heart failure processes such as cardiac hypertrophy, fibrosis, stiffening of arteries, inflammation, and oxidative stress. MR antagonists (MRAs) have shown clear clinical benefits in patients with heart failure with reduced ejection fraction (HFrEF). However, in cases of heart failure with preserved ejection fraction (HFpEF), there is considerable diversity due to its complex underlying mechanisms, resulting in conflicting findings regarding the effectiveness of MRAs in relevant studies. The concept of phenomapping presents an encouraging avenue for investigating different intervention targets and novel therapies for HFpEF. Post hoc analysis of the TOPCAT trial identified certain HFpEF phenotypes that responded favorably to spironolactone. Growing clinical and preclinical evidence suggests that non-steroidal MRAs, which exhibit greater receptor selectivity, stronger anti-fibrotic and anti-inflammatory properties, and fewer hormone-related side effects, may emerge as another promising treatment option for HFpEF alongside sodium-glucose co-transporter 2 (SGLT2) inhibitors. This review aims to outline the structural and functional characteristics of MR, discuss the physiological effects of its activation and inhibition, and delve into the potential for personalized MRA therapy based on the concept of HFpEF phenotype.
Collapse
Affiliation(s)
- Xi Chen
- Department of Cardiology, Affiliated Hospital of Putian University, School of Clinical Medicine, Fujian Medical University, Putian, 351100, China
| | - Meinv Huang
- Department of Cardiology, Affiliated Hospital of Putian University, School of Clinical Medicine, Fujian Medical University, Putian, 351100, China
| | - Yi Chen
- Department of Cardiology, Affiliated Hospital of Putian University, School of Clinical Medicine, Fujian Medical University, Putian, 351100, China
| | - Haishan Xu
- Department of Nephrology, Affiliated Hospital of Putian University, School of Clinical Medicine, Fujian Medical University, Putian, 351100, China.
| | - Meifang Wu
- Department of Cardiology, Affiliated Hospital of Putian University, School of Clinical Medicine, Fujian Medical University, Putian, 351100, China.
| |
Collapse
|
2
|
Daneshpour H, Brüdgam D, Stüfchen I, Heinrich DA, Bidlingmaier M, Beuschlein F, Kürzinger L, Williams TA, Reincke M, Schneider H, Adolf C. Impact of confirmatory test results on subtype classification and biochemical outcome following unilateral adrenalectomy in patients with primary aldosteronism. Front Endocrinol (Lausanne) 2024; 15:1495959. [PMID: 39678193 PMCID: PMC11637841 DOI: 10.3389/fendo.2024.1495959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/05/2024] [Indexed: 12/17/2024] Open
Abstract
Context Primary aldosteronism (PA) is the most common form of endocrine hypertension. According to the Endocrine Society Practice Guidelines, the diagnosis of PA requires a pathological screening test result and non-suppressible aldosterone levels during confirmatory testing. Sequential testing with more than one confirmatory test may result in discordant test results. Objective and patients We investigated the association of discordant results of captopril challenge test (CCT) and saline infusion test (SIT) on patient subtype classification by adrenal vein sampling (AVS) and outcome in 111 consecutive patients from the German Conn's Registry. Concordance was defined as non-suppressible aldosterone levels upon both tests, while discordance was defined as conflicting test results. Patients with unilateral disease were offered adrenalectomy (ADX). Biochemical and clinical outcomes were assessed using the PASO criteria. Results 85 of 111 (77%) patients had concordant results of CCT and SIT. Although baseline characteristics were comparable between patients with concordant and discordant tests, the latter had significantly lower aldosterone levels after testing (CCT: 170 vs. 114pg/ml; SIT: 139 vs. 101pg/ml; p=0.004). In 35% of patients with discordant (n=9) and 46% of concordant test results (n=39), AVS suggested lateralized PA. In 36 of 48 cases ADX was performed. 86% of patients with discordant and 72% with concordant results had complete biochemical success. Conclusion The use of two confirmatory tests in patients with PA results in discordant results in approximately 23% of cases. Patients having discordant confirmatory test results had a comparable rate of lateralized PA and underwent adrenalectomy with similar long-term outcome.
Collapse
Affiliation(s)
- Hediyeh Daneshpour
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Denise Brüdgam
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Isabel Stüfchen
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Daniel Alexander Heinrich
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Martin Bidlingmaier
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich (USZ) und Universität Zürich (UZH), Zurich, Switzerland
- The LOOP Zurich - Medical Research Center, Zurich, Switzerland
| | - Lydia Kürzinger
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital of Würzburg, Würzburg, Germany
| | - Tracy Ann Williams
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Holger Schneider
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Christian Adolf
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| |
Collapse
|
3
|
Ho QV, Young MJ. Mineralocorticoid receptors, macrophages and new mechanisms for cardiovascular disease. Mol Cell Endocrinol 2024; 593:112340. [PMID: 39134137 DOI: 10.1016/j.mce.2024.112340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Affiliation(s)
- Quoc Viet Ho
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Australia
| | - Morag J Young
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Australia; Department of Cardiometabolic Health, University of Melbourne, Victoria, Australia.
| |
Collapse
|
4
|
Hundemer GL, Leung AA, Kline GA, Brown JM, Turcu AF, Vaidya A. Biomarkers to Guide Medical Therapy in Primary Aldosteronism. Endocr Rev 2024; 45:69-94. [PMID: 37439256 PMCID: PMC10765164 DOI: 10.1210/endrev/bnad024] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/23/2023] [Accepted: 07/11/2023] [Indexed: 07/14/2023]
Abstract
Primary aldosteronism (PA) is an endocrinopathy characterized by dysregulated aldosterone production that occurs despite suppression of renin and angiotensin II, and that is non-suppressible by volume and sodium loading. The effectiveness of surgical adrenalectomy for patients with lateralizing PA is characterized by the attenuation of excess aldosterone production leading to blood pressure reduction, correction of hypokalemia, and increases in renin-biomarkers that collectively indicate a reversal of PA pathophysiology and restoration of normal physiology. Even though the vast majority of patients with PA will ultimately be treated medically rather than surgically, there is a lack of guidance on how to optimize medical therapy and on key metrics of success. Herein, we review the evidence justifying approaches to medical management of PA and biomarkers that reflect endocrine principles of restoring normal physiology. We review the current arsenal of medical therapies, including dietary sodium restriction, steroidal and nonsteroidal mineralocorticoid receptor antagonists, epithelial sodium channel inhibitors, and aldosterone synthase inhibitors. It is crucial that clinicians recognize that multimodal medical treatment for PA can be highly effective at reducing the risk for adverse cardiovascular and kidney outcomes when titrated with intention. The key biomarkers reflective of optimized medical therapy are unsurprisingly similar to the physiologic expectations following surgical adrenalectomy: control of blood pressure with the fewest number of antihypertensive agents, normalization of serum potassium without supplementation, and a rise in renin. Pragmatic approaches to achieve these objectives while mitigating adverse effects are reviewed.
Collapse
Affiliation(s)
- Gregory L Hundemer
- Department of Medicine, Division of Nephrology, University of Ottawa, Ottawa, ON K1H 8L6, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Alexander A Leung
- Department of Medicine, Division of Endocrinology and Metabolism, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Gregory A Kline
- Department of Medicine, Division of Endocrinology and Metabolism, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Jenifer M Brown
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Adina F Turcu
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anand Vaidya
- Center for Adrenal Disorders, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
5
|
Smart CD, Madhur MS. The immunology of heart failure with preserved ejection fraction. Clin Sci (Lond) 2023; 137:1225-1247. [PMID: 37606086 PMCID: PMC10959189 DOI: 10.1042/cs20230226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/23/2023] [Accepted: 07/31/2023] [Indexed: 08/23/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) now accounts for the majority of new heart failure diagnoses and continues to increase in prevalence in the United States. Importantly, HFpEF is a highly morbid, heterogeneous syndrome lacking effective therapies. Inflammation has emerged as a potential contributor to the pathogenesis of HFpEF. Many of the risk factors for HFpEF are also associated with chronic inflammation, such as obesity, hypertension, aging, and renal dysfunction. A large amount of preclinical evidence suggests that immune cells and their associated cytokines play important roles in mediating fibrosis, oxidative stress, metabolic derangements, and endothelial dysfunction, all potentially important processes in HFpEF. How inflammation contributes to HFpEF pathogenesis, however, remains poorly understood. Recently, a variety of preclinical models have emerged which may yield much needed insights into the causal relationships between risk factors and the development of HFpEF, including the role of specific immune cell subsets or inflammatory pathways. Here, we review evidence in animal models and humans implicating inflammation as a mediator of HFpEF and identify gaps in knowledge requiring further study. As the understanding between inflammation and HFpEF evolves, it is hoped that a better understanding of the mechanisms underlying immune cell activation in HFpEF can open up new therapeutic avenues.
Collapse
Affiliation(s)
- Charles Duncan Smart
- Department of Molecular Physiology and Biophysics,
Vanderbilt University School of Medicine, Nashville, TN, U.S.A
| | - Meena S. Madhur
- Department of Molecular Physiology and Biophysics,
Vanderbilt University School of Medicine, Nashville, TN, U.S.A
- Department of Medicine, Division of Cardiovascular
Medicine, Vanderbilt University Medical Center, Nashville, TN, U.S.A
- Department of Medicine, Division of Clinical Pharmacology,
Vanderbilt University Medical Center, Nashville, TN, U.S.A
- Vanderbilt Institute for Infection, Immunology, and
Inflammation, Nashville, TN, U.S.A
| |
Collapse
|
6
|
Sotillo S, Ward JL, Guillot E, Domenig O, Yuan L, Smith JS, Gabriel V, Iennarella-Servantez CA, Mochel JP. Dose-response of benazepril on biomarkers of the classical and alternative pathways of the renin-angiotensin-aldosterone system in dogs. Sci Rep 2023; 13:2684. [PMID: 36792677 PMCID: PMC9932142 DOI: 10.1038/s41598-023-29771-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Angiotensin-converting enzyme inhibitors (ACEI) such as benazepril are commonly prescribed in both humans and dogs with heart disease to mitigate the renin-angiotensin-aldosterone system (RAAS); however, the dose-dependent effects of benazepril on comprehensive RAAS components remain unknown. In this study, nine purpose-bred healthy dogs received three different dosages of oral benazepril (0.125 mg/kg, 0.25 mg/kg, or 0.5 mg/kg) in a randomized crossover design following induction of RAAS activation by consuming a low-sodium diet. Blood samples were collected at serial time intervals after benazepril dosing to measure plasma benazeprilat (active metabolite of benazepril) and serum RAAS biomarkers. Blood pressure and echocardiogram were performed at baseline and after each benazepril administration. Time-weighted averages for RAAS biomarkers for 12 h post-dose and hemodynamic variables were compared between dosing groups using Wilcoxon rank-sum testing. Compared to the lowest dosage of benazepril (0.125 mg/kg), the highest dosage (0.5 mg/kg) resulted in lower time-weighted average values of angiotensin (Ang) II (- 38%, P = 0.004), Ang1-5 (- 53%, P = 0.001), ACE-S (surrogate for ACE activity; - 59%, P = 0.0002), and ALT-S (surrogate for alternative RAAS activity; - 22%, P = 0.004), and higher values of AngI (+ 78%, P = 0.014) and PRA-S (surrogate for plasma renin activity; + 58%, P = 0.040). There were no relevant differences between dosing groups for blood pressure or echocardiographic variables. Knowledge of dose-dependent alterations in biomarkers of the classical and alternative RAAS pathways could help inform clinical trials for dosage optimization in both dogs and humans.
Collapse
Affiliation(s)
- Samantha Sotillo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Jessica L Ward
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA.
| | - Emilie Guillot
- Ceva Santé Animale, Companion Animal Franchise, Libourne, France
| | | | - Lingnan Yuan
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Joseph S Smith
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Vojtech Gabriel
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | | | - Jonathan P Mochel
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| |
Collapse
|
7
|
A New Perspective on the Renin-Angiotensin System. Diagnostics (Basel) 2022; 13:diagnostics13010016. [PMID: 36611307 PMCID: PMC9818283 DOI: 10.3390/diagnostics13010016] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in the world. Hypertension is a serious medical problem not only in adults but also in children and adolescents. The renin-angiotensin-aldosterone system (RAAS) is one of the most important mechanisms regulating blood pressure and the balance of water and electrolytes. According to the latest reports, RAAS acts not only on endocrine but also on paracrine, autocrine, and intracrine. Moreover, RAAS has a component associated with hypotension and cardioprotective effects. These components are called alternative pathways of RAAS. The most important peptide of the alternative pathway is Ang 1-7, which is related to the Mas receptor. Mas receptors have widely known antihypertension properties, including vasodilatation, the release of nitric oxide, and increased production of anti-inflammatory cytokines. Another interesting peptide is angiotensin A, which combines the properties of the classical and alternative pathways. No less important components of RAAS are the proteolytic enzymes angiotensin convertase enzyme type 1 and 2. They are responsible for the functioning of the RAAS system and are a hypertension therapeutic target. Also involved are tissue-specific enzymes that form a local renin-angiotensin system. Currently, a combination of drugs is used in hypertension treatment. These drugs have many undesirable side effects that cannot always be avoided. For this reason, new treatments are being sought, and the greatest hope comes from the ACE2/ang 1-7/MasR axis.
Collapse
|
8
|
Roh J, Hill JA, Singh A, Valero-Muñoz M, Sam F. Heart Failure With Preserved Ejection Fraction: Heterogeneous Syndrome, Diverse Preclinical Models. Circ Res 2022; 130:1906-1925. [PMID: 35679364 PMCID: PMC10035274 DOI: 10.1161/circresaha.122.320257] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) represents one of the greatest challenges facing cardiovascular medicine today. Despite being the most common form of heart failure worldwide, there has been limited success in developing therapeutics for this syndrome. This is largely due to our incomplete understanding of the biology driving its systemic pathophysiology and the heterogeneity of clinical phenotypes, which are increasingly being recognized as distinct HFpEF phenogroups. Development of efficacious therapeutics fundamentally relies on robust preclinical models that not only faithfully recapitulate key features of the clinical syndrome but also enable rigorous investigation of putative mechanisms of disease in the context of clinically relevant phenotypes. In this review, we propose a preclinical research strategy that is conceptually grounded in model diversification and aims to better align with our evolving understanding of the heterogeneity of clinical HFpEF. Although heterogeneity is often viewed as a major obstacle in preclinical HFpEF research, we challenge this notion and argue that embracing it may be the key to demystifying its pathobiology. Here, we first provide an overarching guideline for developing HFpEF models through a stepwise approach of comprehensive cardiac and extra-cardiac phenotyping. We then present an overview of currently available models, focused on the 3 leading phenogroups, which are primarily based on aging, cardiometabolic stress, and chronic hypertension. We discuss how well these models reflect their clinically relevant phenogroup and highlight some of the more recent mechanistic insights they are providing into the complex pathophysiology underlying HFpEF.
Collapse
Affiliation(s)
- Jason Roh
- Cardiovascular Research Center, Massachusetts General Hospital, Boston (J.R., A.S.)
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology) (J.A.H.), University of Texas Southwestern Medical Center, Dallas
- Department of Molecular Biology (J.A.H.), University of Texas Southwestern Medical Center, Dallas
| | - Abhilasha Singh
- Cardiovascular Research Center, Massachusetts General Hospital, Boston (J.R., A.S.)
| | - María Valero-Muñoz
- Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (M.V.-M., F.S.)
| | - Flora Sam
- Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (M.V.-M., F.S.)
| |
Collapse
|
9
|
McNally RJ, Farukh B, Chowienczyk PJ, Faconti L. Effect of diuretics on plasma aldosterone and potassium in primary hypertension: A systematic review and meta-analysis. Br J Clin Pharmacol 2022; 88:1964-1977. [PMID: 34820874 DOI: 10.1111/bcp.15156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 11/01/2021] [Accepted: 11/13/2021] [Indexed: 12/18/2022] Open
Abstract
AIM By contrast with drugs inhibiting the renin-angiotensin-aldosterone system (RAAS), diuretics stimulate renin release by the kidneys. Although plasma aldosterone (PA) is thought to be mainly regulated by RAAS activity, serum potassium has been shown to be an important factor in animal models and humans. Here we perform a systematic review and meta-analysis of randomised controlled trials (RCT) in hypertension investigating the effects of diuretic therapy on PA and the correlation of change in PA with that of potassium and blood pressure (BP). METHODS Three databases were searched: MEDLINE, EMBASE and the Cochrane Central Register of Controlled Trials (CENTRAL). Titles were first screened by title and abstract for relevance before full-text articles were assessed for eligibility according to a predefined inclusion/exclusion criteria. RESULTS A total of 1139 articles were retrieved, of which 42 met the prespecified inclusion/exclusion criteria. The average standardised difference in mean PA was similar for all classes of diuretic: thiazide/thiazide-like 0.299 (95% confidence interval [CI] 0.150, 0.447), loop 0.927 (0.37, 1.49), MRA/potassium-sparing 0.265 (0.173, 0.357) and combination 0.466 (0.137, 0.796), Q = 6.33, P = .097. In subjects untreated with another antihypertensive, there was a significant relationship between change in PA and change in systolic BP but no relationship with the change in potassium. CONCLUSION In RCTs of diuretic therapy in hypertension, there is an increase in PA with all classes of diuretic and no significant between-class heterogeneity. Change in PA is not related with potassium but correlates with the change in BP in subjects untreated with another antihypertensive medication.
Collapse
Affiliation(s)
- Ryan J McNally
- Department of Clinical Pharmacology, King's College London British Heart Foundation Centre, London, UK
| | - Bushra Farukh
- Department of Clinical Pharmacology, King's College London British Heart Foundation Centre, London, UK
| | - Philip J Chowienczyk
- Department of Clinical Pharmacology, King's College London British Heart Foundation Centre, London, UK
| | - Luca Faconti
- Department of Clinical Pharmacology, King's College London British Heart Foundation Centre, London, UK
| |
Collapse
|
10
|
Bauersachs J, Lother A. Mineralocorticoid receptor activation and antagonism in cardiovascular disease: cellular and molecular mechanisms. Kidney Int Suppl (2011) 2022; 12:19-26. [DOI: 10.1016/j.kisu.2021.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 02/08/2023] Open
|
11
|
Yu S, Ren J, Lv Z, Li R, Zhong Y, Yao W, Yuan J. Prediction of the endocrine-disrupting ability of 49 per- and polyfluoroalkyl substances: In silico and epidemiological evidence. CHEMOSPHERE 2022; 290:133366. [PMID: 34933031 DOI: 10.1016/j.chemosphere.2021.133366] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 06/14/2023]
Abstract
The toxic effects of per- and polyfluoroalkyl substances (PFASs) on humans are mediated by nuclear hormone receptors (NHRs). However, data on the interaction of PFASs and NHRs is limited. Endocrine Disruptome, an inverse docking tool, was used in this study to simulate the docking of 49 common PFASs with 14 different types of human NHRs. According to the findings, 25 PFASs have a high or moderately high probability of binding to more than five NHRs, with androgen receptor (AR) and mineralocorticoid receptor (MR) being the most likely target NHRs. Molecular docking analyses revealed that the binding modes of PFASs with the two NHRs were similar to those of their corresponding co-crystallized ligands. PFASs, in particular, may disrupt the endocrine system by binding to MR. This finding is consistent with epidemiological research that has linked PFASs to MR-related diseases. Our findings may contribute to a better understanding of the health risks posed by PFASs.
Collapse
Affiliation(s)
- Shuling Yu
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, Henan, 475004, PR China
| | - Jing Ren
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Zhenxia Lv
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Rui Li
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Yuyan Zhong
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Wu Yao
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Jintao Yuan
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China.
| |
Collapse
|
12
|
Serum collected from rats with myocardial infarction increases extracellular matrix accumulation by myofibroblasts isolated from myocardial infarction scar. EUROBIOTECH JOURNAL 2022. [DOI: 10.2478/ebtj-2022-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
The effect on extracellular matrix content is believed to be an average of several serum derived compounds acting in opposition. The aim of the study is to determine whether whole serum of rats with myocardial infarction may modify the accumulation of extracellular matrix in cultures of myofibroblasts isolated from the myocardial infarction scar. A second aim is to determine whether the tested serum can also degranulate the mast cells. Serum was collected from rats with sham myocardial infarction, rats with myocardial infarction induced by coronary artery ligation and control animals. The experiments were carried out on myocardial infarction scar myofibroblasts or mast cells from the peritoneal cavity. The cultures were divided into three groups containing eight cultures each: one treated with serum from control rats, from animals after sham operation or from those after myocardial infarction. In all groups, the serum was used at concentrations of 10%, 20% or 30%. The total collagen content (Woesner method) glycosaminoglycan level (Farandale method), cell proliferation (BrdU), histamine secretion from mast cells (spectrofluorymetry), β1 integrin and α-smooth muscle actin expression (flow cytometry) were evaluated. Isolated cells were α-smooth muscle actin positive and identified as myofibroblasts. Serum derived from rats with myocardial infarction increased collagen and glycosaminoglycan content in the cultures and modified myofibroblast proliferation in a concentration-dependent manner. The serum also results in an imbalance between collagen and glycosaminoglycan levels. The content of β1 integrin was not influenced by myocardial infarction serum. The serum of rats with myocardial infarction is involved in regulation of collagen and glycosaminoglycan content in myofibroblast cultures, as well as the modification of their proliferation. These changes were not accompanied with integrin β1 density variations. The serum of the myocardial infarction rats did not influence the mast cell degranulation.
Collapse
|
13
|
Impact of Loop Diuretic on Outcomes in Patients with Heart Failure and Reduced Ejection Fraction. Curr Heart Fail Rep 2022; 19:15-25. [PMID: 35037162 DOI: 10.1007/s11897-021-00538-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/14/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Loop diuretics are the cornerstone of the treatment of congestion in heart failure patients. The manuscript aims to summarize the most updated information regarding the use of loop diuretics in heart failure. RECENT FINDINGS Diuretic response can be highly variable between patients and needs to be carefully evaluated during and after the hospitalization. Diuretic resistance can lead to residual congestion which affects prognosis and can be difficult to detect. The effect of loop diuretics on long-term prognosis remains uncertain but patients with advanced heart failure typically have renal dysfunction and are more inclined to develop loop diuretic resistance, which may lead to an incomplete decongestion and thus to a worse prognosis. Loop diuretics are the most potent diuretics available and their use is recommended in order to alleviate symptoms, improve exercise capacity, and reduce hospitalizations in patients with heart failure. Their use should be limited to the lowest dose necessary to maintain euvolemia because a low dose does not increase the risk of decompensation but reduce the risk of adverse effects and allow the up-titration of disease-modifying drugs.
Collapse
|
14
|
Grossmann C, Almeida-Prieto B, Nolze A, Alvarez de la Rosa D. Structural and molecular determinants of mineralocorticoid receptor signalling. Br J Pharmacol 2021; 179:3103-3118. [PMID: 34811739 DOI: 10.1111/bph.15746] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/19/2021] [Accepted: 11/08/2021] [Indexed: 12/18/2022] Open
Abstract
During the past decades, the mineralocorticoid receptor (MR) has evolved from a much-overlooked member of the steroid hormone receptor family to an important player, not only in volume and electrolyte homeostasis but also in pathological changes occurring in an increasing number of tissues, especially the renal and cardiovascular systems. Simultaneously, a wealth of information about the structure, interaction partners and chromatin requirements for genomic signalling of steroid hormone receptors became available. However, much of the information for the MR has been deduced from studies of other family members and there is still a lack of knowledge about MR-specific features in ligand binding, chromatin remodelling, co-factor interactions and general MR specificity-conferring mechanisms that can completely explain the differences in pathophysiological function between MR and its closest relative, the glucocorticoid receptor. This review aims to give an overview of the current knowledge of MR structure, signalling and co-factors modulating its activity.
Collapse
Affiliation(s)
- Claudia Grossmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle, Saale, Germany
| | - Brian Almeida-Prieto
- Departamento de Ciencias Médicas Básicas and Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Alexander Nolze
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle, Saale, Germany
| | - Diego Alvarez de la Rosa
- Departamento de Ciencias Médicas Básicas and Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, Tenerife, Spain
| |
Collapse
|
15
|
Effect of rs4646994 polymorphism of angiotensin converting enzyme on the risk of nonischemic cardiomyopathy. Biosci Rep 2021; 41:230176. [PMID: 34750628 PMCID: PMC8685642 DOI: 10.1042/bsr20211617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/12/2021] [Accepted: 10/27/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Angiotensin-converting enzyme (ACE) gene polymorphisms have recently been shown to be associated with risk of developing left ventricular hypertrophy (LVH). However, the results were controversial. We aimed to conduct this meta-analysis to further confirm the association between ACE rs4646994 polymorphism and hypertrophic cardiomyopathy (HCM)/dilated cardiomyopathy (DCM). Methods: PubMed, Embase, the Chinese National Knowledge Information, and Wanfang databases were searched for eligible studies. The Newcastle–Ottawa Scale (NOS) was used to evaluate the quality of included studies. Then we evaluated the association between ACE gene mutation and HCM/DCM by calculating odds ratios (ORs) and 95% confidence intervals (95% CIs). Subgroup analysis was further performed to explore situations in specialized subjects. Sensitivity analysis and publication bias was assessed to confirm the study reliability. Results: There were 13 studies on DCM (2004 cases and 1376 controls) and 16 studies on HCM (2161 controls and 1192 patients). ACE rs4646994 polymorphism was significantly associated with DCM in all genetic models. However, in HCM, four genetic models (allele model, homozygous model, heterozygous model, and dominant model) showed significant association between ACE rs4646994 polymorphism and DCM. In subgroup analysis, we found that ACE rs4646994 polymorphism was significantly associated with DCM/HCM in Asian population. Finally, we also conducted a cumulative meta-analysis, which indicates that the results of our meta-analysis are highly reliable. Conclusion: ACE rs4646994 polymorphism increases the risk of DCM/HCM in Asians, but not in Caucasians. More case–control studies are needed to strengthen our conclusions and to assess the gene–gene and gene–environment interactions between ACE rs4646994 polymorphism and DCM/HCM.
Collapse
|
16
|
Young MJ, Kanki M, Karthigan N, Konstandopoulos P. The Role of the Mineralocorticoid Receptor and Mineralocorticoid Receptor-Directed Therapies in Heart Failure. Endocrinology 2021; 162:6288445. [PMID: 34050730 DOI: 10.1210/endocr/bqab105] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mineralocorticoid receptor (MR) antagonists (MRA), also referred to as aldosterone blockers, are now well-recognized for their clinical benefit in patients who have heart failure (HF) with reduced ejection fraction (HFrEF). Recent studies have also shown MRA can improve outcomes in patients with HFpEF, where the ejection fraction is preserved but left ventricular filling is reduced. While the MR is a steroid hormone receptor best known for antinatriuretic actions on electrolyte homeostasis in the distal nephron, it is now established that the MR has many physiological and pathophysiological roles in the heart, vasculature, and other nonepithelial tissue types. It is the impact of MR activation on these tissues that underpins the use of MRA in cardiovascular disease, in particular HF. This mini-review will discuss the origins and the development of MRA and highlight how their use has evolved from the "potassium-sparing diuretics" spironolactone and canrenone over 60 years ago, to the more receptor-selective eplerenone and most recently the emergence of new nonsteroidal receptor antagonists esaxerenone and finerenone.
Collapse
Affiliation(s)
- Morag J Young
- Baker Heart and Diabetes Institute, Cardiovascular Endocrinology Laboratory, Prahran 3181, Australia
| | - Monica Kanki
- Baker Heart and Diabetes Institute, Cardiovascular Endocrinology Laboratory, Prahran 3181, Australia
- Hudson Institute of Medical Research, Victoria 3168, Australia
| | - Nikshay Karthigan
- Baker Heart and Diabetes Institute, Cardiovascular Endocrinology Laboratory, Prahran 3181, Australia
- Hudson Institute of Medical Research, Victoria 3168, Australia
| | - Penny Konstandopoulos
- Baker Heart and Diabetes Institute, Cardiovascular Endocrinology Laboratory, Prahran 3181, Australia
| |
Collapse
|
17
|
Xing Q, Wu M, Chen R, Liang G, Duan H, Li S, Wang Y, Wang L, An C, Qin G, Sang N. Comparative studies on regional variations in PM 2.5 in the induction of myocardial hypertrophy in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 775:145179. [PMID: 33611177 DOI: 10.1016/j.scitotenv.2021.145179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/02/2021] [Accepted: 01/12/2021] [Indexed: 06/12/2023]
Abstract
Exposure to fine particulate matter (PM2.5) has been indicated to be related to an increased risk of cardiovascular diseases (CVDs) in sensitive people. However, the underlying mechanisms of PM2.5-induced CVDs are poorly understood. In the present study, PM2.5 samples were collected during winter from four cities (Taiyuan, Beijing, Hangzhou, and Guangzhou) in China. Ten-month-old C57BL/6 female mice were exposed to PM2.5 suspension at a dosage of 3 mg·kg-1 (b. w.) every other day for 4 weeks by oropharyngeal aspiration. PM2.5 from Taiyuan increased the blood pressure and the thicknesses of the left ventricular anterior and posterior walls, decreased the ratio of nucleus to cytoplasm in cardiomyocytes and reduced the systolic function of the heart in mice. Further investigation revealed that PM2.5 from Taiyuan induced lung inflammatory cytokines with up-regulated expressions of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). The mRNA expression levels of myocardial hypertrophy markers atrial natriuretic peptide and the β isoform of myosin heavy chain (ANP and β-MHC), matrix metalloproteinase 2 (MMP2), MMP9, and inflammatory cytokines TNF-α and IL-6 in the myocardium were significantly increased after exposure to PM2.5 of Taiyuan. Furthermore, PM2.5 from Taiyuan activated the IL-6/JAK2/STAT3/β-MHC signaling pathway in the myocardium. The correlation between the PM2.5 components and myocardial hypertrophy markers suggested that Zinc (Zn) and acenaphthene (AC) are related to the changes in ANP and β-MHC at the transcriptional level, respectively. The above results indicated that PM2.5 exposure induced myocardial hypertrophy in older mice, which might be related to the critical contributions of Zn and AC in PM2.5. The present study provides new insights into the mechanism of myocardial hypertrophy after PM2.5 exposure.
Collapse
Affiliation(s)
- Qisong Xing
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Meiqiong Wu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China; School of Public Health, Shanxi Medical University, Shanxi 030001, PR China
| | - Rui Chen
- Beijing Key Laboratory of Occupational Safety and Health, Beijing Municipal Institute of Labour Protection, Beijing Academy of Science and Technology, Beijing 100054, PR China
| | - Gang Liang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Huiling Duan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Shuyue Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Yuqian Wang
- Beijing Key Laboratory of Occupational Safety and Health, Beijing Municipal Institute of Labour Protection, Beijing Academy of Science and Technology, Beijing 100054, PR China
| | - Lei Wang
- Key laboratory of Mineral Resources and Ecological Environment Monitoring, Hebei Research Center for Geoanalysis, Baoding, Hebei 071000, PR China
| | - Caixiu An
- Key laboratory of Mineral Resources and Ecological Environment Monitoring, Hebei Research Center for Geoanalysis, Baoding, Hebei 071000, PR China
| | - Guohua Qin
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| |
Collapse
|
18
|
Adolf C, Görge V, Heinrich DA, Hoster E, Schneider H, Handgriff L, Künzel H, Sturm L, Beuschlein F, Reincke M. Altered Taste Perception for Sodium Chloride in Patients With Primary Aldosteronism: A Prospective Cohort Study. Hypertension 2021; 77:1332-1340. [PMID: 33641355 DOI: 10.1161/hypertensionaha.120.16440] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Christian Adolf
- From the Medizinische Klinik und Poliklinik IV (C.A., V.G., D.A.H., H.S., L.H., H.K., L.S., F.B., M.R.), Klinikum der Universität München, LMU München, Munich, Germany
| | - Veronika Görge
- From the Medizinische Klinik und Poliklinik IV (C.A., V.G., D.A.H., H.S., L.H., H.K., L.S., F.B., M.R.), Klinikum der Universität München, LMU München, Munich, Germany
| | - Daniel A Heinrich
- From the Medizinische Klinik und Poliklinik IV (C.A., V.G., D.A.H., H.S., L.H., H.K., L.S., F.B., M.R.), Klinikum der Universität München, LMU München, Munich, Germany
| | - Eva Hoster
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE) (E.H.), Klinikum der Universität München, LMU München, Munich, Germany.,Department of Medicine III (E.H.), Klinikum der Universität München, LMU München, Munich, Germany
| | - Holger Schneider
- From the Medizinische Klinik und Poliklinik IV (C.A., V.G., D.A.H., H.S., L.H., H.K., L.S., F.B., M.R.), Klinikum der Universität München, LMU München, Munich, Germany
| | - Laura Handgriff
- From the Medizinische Klinik und Poliklinik IV (C.A., V.G., D.A.H., H.S., L.H., H.K., L.S., F.B., M.R.), Klinikum der Universität München, LMU München, Munich, Germany
| | - Heike Künzel
- From the Medizinische Klinik und Poliklinik IV (C.A., V.G., D.A.H., H.S., L.H., H.K., L.S., F.B., M.R.), Klinikum der Universität München, LMU München, Munich, Germany
| | - Lisa Sturm
- From the Medizinische Klinik und Poliklinik IV (C.A., V.G., D.A.H., H.S., L.H., H.K., L.S., F.B., M.R.), Klinikum der Universität München, LMU München, Munich, Germany
| | - Felix Beuschlein
- From the Medizinische Klinik und Poliklinik IV (C.A., V.G., D.A.H., H.S., L.H., H.K., L.S., F.B., M.R.), Klinikum der Universität München, LMU München, Munich, Germany.,Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Switzerland (F.B.)
| | - Martin Reincke
- From the Medizinische Klinik und Poliklinik IV (C.A., V.G., D.A.H., H.S., L.H., H.K., L.S., F.B., M.R.), Klinikum der Universität München, LMU München, Munich, Germany
| |
Collapse
|
19
|
Lottspeich C, Köhler A, Czihal M, Heinrich DA, Schneider H, Handgriff L, Reincke M, Adolf C. Atherosclerotic Burden and Arterial Stiffness are Not Increased in Patients with Milder Forms of Primary Aldosteronism Compared to Patients with Essential Hypertension. Horm Metab Res 2021; 53:178-184. [PMID: 33440431 PMCID: PMC7924991 DOI: 10.1055/a-1326-2164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Patients with primary aldosteronism (PA) are at increased cardiovascular risk, compared to patients with essential hypertension (EH). Cardiovascular damage could depend on PA phenotype, potentially being lower in milder forms of PA. Our aim was to assess atherosclerotic burden and arterial stiffness in 88 prospectively recruited patients, including 44 patients with mild PA and EH respectively. All patients underwent a structured study program, including measurements of ankle-brachial index, oscillometric measurement of central pulse wave velocity (cPWV) and vascular ultrasound examination of the supraaortic arteries, the abdominal aorta, and the femoropopliteal arteries. A plaque score was calculated to estimate atherosclerotic burden for each patient. This is a prospective case-control study set at a tertiary care hospital. Patients with PA and EH matched well for age, gender, blood pressure, BMI, and cardiovascular risk factors such as diabetes mellitus and smoking status. Common carotid intima-media thickness (0.77 vs. 0.75 mm; p=0.997) and cPWV (7.2 vs. 7.1 m/s; p=0.372) were comparable between patients with PA and EH. The atherosclerotic burden, as expressed by the plaque score, did not differ between the two groups (p=0.159). However, after initiation of treatment cPWV was significantly decreased in patients with PA (p=0.017). This study shows that subclinical atherosclerotic burden and arterial stiffness in patients with milder forms of PA is comparable to patients with EH. Nevertheless, specific treatment for PA significantly improved cPWV, which argues for a more liberal use of mineralocorticoid receptor antagonists in patients with arterial hypertension.
Collapse
Affiliation(s)
| | - Anton Köhler
- Medizinische Klinik und Poliklinik IV, LMU München, Munich,
Germany
| | - Michael Czihal
- Medizinische Klinik und Poliklinik IV, LMU München, Munich,
Germany
| | | | - Holger Schneider
- Medizinische Klinik und Poliklinik IV, LMU München, Munich,
Germany
| | - Laura Handgriff
- Medizinische Klinik und Poliklinik IV, LMU München, Munich,
Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, LMU München, Munich,
Germany
| | - Christian Adolf
- Medizinische Klinik und Poliklinik IV, LMU München, Munich,
Germany
- Correspondence Dr. Christian Adolf Medizinische Klinik und Poliklinik IVKlinikum der Universität MünchenZiemssenstr. 180336 MunichGermany+49 89 4400 52458+49 89 4400 52194
| |
Collapse
|
20
|
Young MJ, Kanki M, Fuller PJ, Yang J. Identifying new cellular mechanisms of mineralocorticoid receptor activation in the heart. J Hum Hypertens 2021; 35:124-130. [PMID: 32733061 DOI: 10.1038/s41371-020-0386-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/01/2020] [Accepted: 07/16/2020] [Indexed: 01/30/2023]
Abstract
Recent studies have expanded our understanding of the actions of the mineralocorticoid receptor (MR) to a diverse array of tissue types that differ substantially from the epithelial cells of the renal nephron. In these cell types the role of the MR has been largely, but not exclusively, defined in terms of pathogenic signalling pathways leading to tissue injury and remodelling. Macrophages and cardiomyocytes are two cell types in which the MR plays a central role in the cardiac tissue response to injury, renovascular hypertension and oxidative stress for example. Macrophages are critical for resolution of tissue injury and wound healing and their pleiotropic actions are central to the development of many forms of heart, renal and vascular disease. The MR in cardiomyocytes is not only essential for the chronotropic and ionotropic actions of mineralocorticoids in the short and longer term, but also for induction of hypertrophic and proinflammatory signalling programs. The present review discusses recent studies, presented at the Aldosterone and Hypertension Satellite of the 15th Asian-Pacific Congress of Hypertension, investigating new mechanisms for MR signalling in these cells and how their dysfunction contributes to the onset and progression of cardiovascular disease and heart failure.
Collapse
Affiliation(s)
- Morag J Young
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and the Department of Molecular Translational Science, Monash University, Clayton, VIC, Australia. .,Baker Heart and Diabetes Institute, Melborne, VIC, Australia.
| | - Monica Kanki
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and the Department of Molecular Translational Science, Monash University, Clayton, VIC, Australia.,Baker Heart and Diabetes Institute, Melborne, VIC, Australia
| | - Peter J Fuller
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and the Department of Molecular Translational Science, Monash University, Clayton, VIC, Australia
| | - Jun Yang
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and the Department of Molecular Translational Science, Monash University, Clayton, VIC, Australia
| |
Collapse
|
21
|
Zimmerman AD, Mackay L, Kemppainen RJ, Jones MA, Read CC, Schwartz D, Foradori CD. The Herbicide Atrazine Potentiates Angiotensin II-Induced Aldosterone Synthesis and Release From Adrenal Cells. Front Endocrinol (Lausanne) 2021; 12:697505. [PMID: 34335472 PMCID: PMC8317615 DOI: 10.3389/fendo.2021.697505] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/22/2021] [Indexed: 12/06/2022] Open
Abstract
Atrazine is one of the most commonly used pre-emergence and early post-emergence herbicides in the world. We have shown previously that atrazine does not directly stimulate the pituitary or adrenal to trigger hormone release but acts centrally to activate a stress-like activation of the hypothalamic-pituitary-adrenal axis. In doing so, atrazine treatment has been shown to cause adrenal morphology changes characteristic of repeated stress. In this study, adrenals from atrazine treated and stressed animals were directly compared after 4 days of atrazine treatment or restraint stress. Both atrazine and stressed animals displayed reduced adrenocortical zona glomerulosa thickness and aldosterone synthase (CYP11B2) expression, indicative of repeated adrenal stimulation by adrenocorticotropic hormone. To determine if reduced CYP11B2 expression resulted in attenuated aldosterone synthesis, stressed and atrazine treated animals were challenged with angiotensin II (Ang II). As predicted, stressed animals produced less aldosterone compared to control animals when stimulated. However, atrazine treated animals had higher circulating aldosterone concentrations compared to both stressed and control groups. Ang II-induced aldosterone release was also potentiated in atrazine pretreated human adrenocortical carcinoma cells (H295R). Atrazine pretreated did not alter the expression of the rate limiting steroidogenic StAR protein or angiotensin II receptor 1. Atrazine treated animals also presented with higher basal blood pressure than vehicle treated control animals suggesting sustained elevations in circulating aldosterone levels. Our results demonstrate that treatment with the widely used herbicide, atrazine, directly increases stimulated production of aldosterone in adrenocortical cells independent of expression changes to rate limiting steroidogenic enzymes.
Collapse
|
22
|
Higuchi S, Ota H, Tezuka Y, Seiji K, Takagi H, Lee J, Lee YW, Omata K, Ono Y, Morimoto R, Kudo M, Satoh F, Takase K. Aldosterone-induced cardiac damage in primary aldosteronism depends on its subtypes. Endocr Connect 2021; 10:29-36. [PMID: 33268573 PMCID: PMC7923132 DOI: 10.1530/ec-20-0504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/25/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVES This study compared cardiac function, morphology, and tissue characteristics between two common subtypes of primary aldosteronism (PA) using a 3T MR scanner. DESIGN A retrospective, single-center, observational study. METHODS We retrospectively reviewed 143 consecutive patients with PA, who underwent both adrenal venous sampling and cardiac magnetic resonance. We acquired cine, late gadolinium enhancement, and pre- and postcontrast myocardial T1-mapping images. RESULTS PA was diagnosed as unilateral aldosterone-producing adenoma (APA) in 70 patients and bilateral hyperaldosteronism (BHA) in 73. The APA group showed significantly higher plasma aldosterone concentration (PAC) and aldosterone to renin rate (ARR) than the BHA group. After controlling for age, sex, antihypertensive drugs, systolic and diastolic blood pressure, and disease duration, the parameters independently associated with APA were: left ventricular end-diastolic volume index (EDVI: adjusted odds ratio (aOR) = 1.06 (95% CI: 1.030-1.096), P < 0.01), end-systolic volume index (ESVI: 1.06 (1.017-1.113), P < 0.01), stroke index (SI: 1.07 (1.020-1.121), P < 0.01), cardiac index (CI: 1.001 (1.000-1.001), P < 0.01), and native T1 (1.01 (1.000-1.019), P = 0.038). Weak positive correlations were found between PAC and EDVI (R = 0.28, P < 0.01), ESVI (0.26, P < 0.01), and SI (0.18, P = 0.03); and between ARR and EDVI (0.25, P < 0.01), ESVI (0.24, P < 0.01), and native T1 (0.17, P = 0.047). CONCLUSIONS APA is associated with greater LV volumetric parameters and higher native T1 values, suggesting a higher risk of volume overload and myocardial damage.
Collapse
Affiliation(s)
- Satoshi Higuchi
- Department of Diagnostic Radiology, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Hideki Ota
- Department of Diagnostic Radiology, Tohoku University Hospital, Sendai, Miyagi, Japan
- Department of Advanced MRI Collaboration Research, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Correspondence should be addressed to H Ota:
| | - Yuta Tezuka
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Department of Radiology, The University of British Columbia, Vancouver, Canada
| | - Kazumasa Seiji
- Department of Diagnostic Radiology, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Hidenobu Takagi
- Department of Diagnostic Radiology, Tohoku University Hospital, Sendai, Miyagi, Japan
- Department of Radiology, The University of British Columbia, Vancouver, Canada
| | - Jongmin Lee
- Department of Radiology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Yi-Wei Lee
- Department of Radiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kei Omata
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Department of Radiology, The University of British Columbia, Vancouver, Canada
| | - Yoshikiyo Ono
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Department of Radiology, The University of British Columbia, Vancouver, Canada
| | - Ryo Morimoto
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Masataka Kudo
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Fumitoshi Satoh
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kei Takase
- Department of Diagnostic Radiology, Tohoku University Hospital, Sendai, Miyagi, Japan
| |
Collapse
|
23
|
Song X, Zhao Y, Wang S, Wang Y, Chen Q, Zhao H, Wang H, Tian S, Yu H, Wu Z. Zi Shen Huo Luo Formula Enhances the Therapeutic Effects of Angiotensin-Converting Enzyme Inhibitors on Hypertensive Left Ventricular Hypertrophy by Interfering With Aldosterone Breakthrough and Affecting Caveolin-1/Mineralocorticoid Receptor Colocalization and Downstream Extracellular Signal-Regulated Kinase Signaling. Front Pharmacol 2020; 11:383. [PMID: 32317965 PMCID: PMC7147343 DOI: 10.3389/fphar.2020.00383] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/12/2020] [Indexed: 12/24/2022] Open
Abstract
Left ventricular hypertrophy (LVH) is an important characteristic of hypertensive heart disease. Renin-angiotensin system (RAS) blockers have been shown to be effective drugs for the reversal of LVH. Clinical and experimental studies have shown that Zi Shen Huo Luo Formula (ZSHLF) can improve the efficacy of perindopril in the treatment of hypertensive LVH, but its mechanism is unclear. This study aimed to investigate the possible mechanism to improve the efficacy of perindopril. First, we identified 23 compounds in ZSHLF by ultra performance liquid chromatography/tandem mass spectrometry (UPLC-MS/MS) analysis, among which ferulic acid, caffeic acid, vanillic acid, berberine, rutin, quercetin, kaempferol, stachydrine, and tiliroside have been reported to reduce blood pressure and exhibit cardioprotective effects. Second, we treated spontaneously hypertensive rats (SHRs) with perindopril and ZSHLF for 12 continuous weeks and found that chronic use of perindopril could increase the aldosterone (ALD) levels and cause aldosterone breakthrough (ABT). ZSHLF combined with perindopril reduced the ALD levels, interfered with ABT, decreased blood pressure, improved left ventricular diastolic dysfunction, and decreased the collagen volume fraction; these effects were superior to those of perindopril alone. In vitro experiments, ALD-induced cardiomyocytes (H9c2 cells) and cardiac fibroblasts were treated with ZSHLF-containing serum, which suppressed ALD-induced cardiomyocyte hypertrophy and cardiac fibroblast proliferation, increased mineralocorticoid receptor (MR) and Cav-1 colocalization and decreased phosphorylated epidermal growth factor receptor (pEGFR) and phosphorylated extracellular signal-regulated kinase (pERK) protein expression the cells. In conclusion, ZSHLF can interfere with ABT and affect the pathological role of ALD by affecting MR and Cav-1 interactions and EGFR/ERK signaling pathway. These effects represent a possible mechanism by which ZSHLF improves the efficacy of angiotensin-converting enzyme inhibitors (ACEIs) in hypertensive LVH treatment. However, the major bioactive components or metabolites responsible for the effects and the implications of these findings in patients need further verification.
Collapse
Affiliation(s)
- Xiaotong Song
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Co-innovation Center of Classic TCM Formula, Shandong Provincial Education Department, Jinan, China
| | - Yue Zhao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shijun Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Co-innovation Center of Classic TCM Formula, Shandong Provincial Education Department, Jinan, China
| | - Yuan Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Co-innovation Center of Classic TCM Formula, Shandong Provincial Education Department, Jinan, China
| | - Qian Chen
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Co-innovation Center of Classic TCM Formula, Shandong Provincial Education Department, Jinan, China
| | - Haijun Zhao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Co-innovation Center of Classic TCM Formula, Shandong Provincial Education Department, Jinan, China
| | - Hua Wang
- Department of Geriatric Medicine, Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Sheng Tian
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Co-innovation Center of Classic TCM Formula, Shandong Provincial Education Department, Jinan, China
| | - Huayun Yu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Co-innovation Center of Classic TCM Formula, Shandong Provincial Education Department, Jinan, China
| | - Zhichun Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Co-innovation Center of Classic TCM Formula, Shandong Provincial Education Department, Jinan, China
| |
Collapse
|
24
|
Ersoy R, Celik A, Yilmaz O, Sarioglu S, Sis B, Akan P, Yenicerioglu Y, Ormen M, Camsari T. The Effects of Irbesartan and Spironolactone in Prevention of Peritoneal Fibrosis in Rats. Perit Dial Int 2020. [DOI: 10.1177/089686080702700410] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BackgroundBacterial peritonitis episodes may disturb the functional and histological integrity of the peritoneum in peritoneal dialysis patients. The renin–angiotensin–aldosterone system may have fibrotic effects on the peritoneum.ObjectiveTo study the effects of an angiotensin II receptor antagonist (irbesartan) and an aldosterone antagonist (spironolactone) in the prevention of peritoneal fibrosis in a rat model of bacterial peritonitis.Methods40 Wistar rats were randomized into 5 groups: bacteria (B), bacteria–irbesartan (BI), bacteria–spironolactone (BS), bacteria–irbesartan–spironolactone (BIS), and control (C) groups. The C group received only dextran beads (Cytodex; Sigma Chemicals, St Louis, Missouri, USA); the others were given bacteria and dextran beads intraperitoneally. Irbesartan and/or spironolactone were given to 3 groups: BI, BS, and BIS. On the eighth day, the rats were sacrificed, peritoneal adhesion was quantified, and peritoneal tissue sections were evaluated histologically.ResultsThe peritoneal total adhesion score was significantly higher in the B group than in the BI, BIS, and C groups ( p < 0.01). Mean peritoneal thickness, mean inflammation score, and mean fibrosis score were significantly higher in the B group in comparison to the C group ( p < 0.05). Mean peritoneal thickness of all treatment groups was significantly lower than the B group ( p < 0.05). Serum transforming growth factor beta-1 level was significantly higher in the B group than in the BI, BS, and C groups ( p < 0.05).ConclusionIrbesartan and spironolactone seem to decrease the extent of peritoneal injury caused by bacterial peritonitis.
Collapse
Affiliation(s)
- Rifki Ersoy
- Departments of Nephrology, Dokuz Eylül University Medical School, Izmir, Turkey
| | - Ali Celik
- Departments of Nephrology, Dokuz Eylül University Medical School, Izmir, Turkey
| | - Osman Yilmaz
- Animal Laboratory, Dokuz Eylül University Medical School, Izmir, Turkey
| | - Sulen Sarioglu
- Pathology, Dokuz Eylül University Medical School, Izmir, Turkey
| | - Banu Sis
- Pathology, Dokuz Eylül University Medical School, Izmir, Turkey
| | - Pinar Akan
- Biochemistry, Dokuz Eylül University Medical School, Izmir, Turkey
| | - Yavuz Yenicerioglu
- Departments of Nephrology, Dokuz Eylül University Medical School, Izmir, Turkey
| | - Murat Ormen
- Biochemistry, Dokuz Eylül University Medical School, Izmir, Turkey
| | - Taner Camsari
- Departments of Nephrology, Dokuz Eylül University Medical School, Izmir, Turkey
| |
Collapse
|
25
|
Chen ZW, Tsai CH, Pan CT, Chou CH, Liao CW, Hung CS, Wu VC, Lin YH. Endothelial Dysfunction in Primary Aldosteronism. Int J Mol Sci 2019; 20:ijms20205214. [PMID: 31640178 PMCID: PMC6829211 DOI: 10.3390/ijms20205214] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Primary aldosteronism (PA) is characterized by excess production of aldosterone from the adrenal glands and is the most common and treatable cause of secondary hypertension. Aldosterone is a mineralocorticoid hormone that participates in the regulation of electrolyte balance, blood pressure, and tissue remodeling. The excess of aldosterone caused by PA results in an increase in cardiovascular and cerebrovascular complications, including coronary artery disease, myocardial infarction, stroke, transient ischemic attack, and even arrhythmia and heart failure. Endothelial dysfunction is a well-established fundamental cause of cardiovascular diseases and also a predictor of worse clinical outcomes. Accumulating evidence indicates that aldosterone plays an important role in the initiation and progression of endothelial dysfunction. Several mechanisms have been shown to contribute to aldosterone-induced endothelial dysfunction, including aldosterone-mediated vascular tone dysfunction, aldosterone- and endothelium-mediated vascular inflammation, aldosterone-related atherosclerosis, and vascular remodeling. These mechanisms are activated by aldosterone through genomic and nongenomic pathways in mineralocorticoid receptor-dependent and independent manners. In addition, other cells have also been shown to participate in these mechanisms. The complex interactions among endothelium, inflammatory cells, vascular smooth muscle cells and fibroblasts are crucial for aldosterone-mediated endothelial dysregulation. In this review, we discuss the association between aldosterone and endothelial function and the complex mechanisms from a molecular aspect. Furthermore, we also review current clinical research of endothelial dysfunction in patients with PA.
Collapse
Affiliation(s)
- Zheng-Wei Chen
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan.
- Cardiovascular center, National Taiwan University Hospital, Taipei 10002, Taiwan.
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin 64041, Taiwan.
| | - Cheng-Hsuan Tsai
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan.
- Cardiovascular center, National Taiwan University Hospital, Taipei 10002, Taiwan.
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Jin-Shan Branch, New Taipei City 20844, Taiwan.
| | - Chien-Ting Pan
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan.
- Cardiovascular center, National Taiwan University Hospital, Taipei 10002, Taiwan.
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin 64041, Taiwan.
| | - Chia-Hung Chou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10041, Taiwan.
| | - Che-Wei Liao
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu 30059, Taiwan.
| | - Chi-Sheng Hung
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan.
- Cardiovascular center, National Taiwan University Hospital, Taipei 10002, Taiwan.
| | - Vin-Cent Wu
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan.
| | - Yen-Hung Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan.
- Cardiovascular center, National Taiwan University Hospital, Taipei 10002, Taiwan.
| |
Collapse
|
26
|
Tsutsui H, Isobe M, Ito H, Ito H, Okumura K, Ono M, Kitakaze M, Kinugawa K, Kihara Y, Goto Y, Komuro I, Saiki Y, Saito Y, Sakata Y, Sato N, Sawa Y, Shiose A, Shimizu W, Shimokawa H, Seino Y, Node K, Higo T, Hirayama A, Makaya M, Masuyama T, Murohara T, Momomura SI, Yano M, Yamazaki K, Yamamoto K, Yoshikawa T, Yoshimura M, Akiyama M, Anzai T, Ishihara S, Inomata T, Imamura T, Iwasaki YK, Ohtani T, Onishi K, Kasai T, Kato M, Kawai M, Kinugasa Y, Kinugawa S, Kuratani T, Kobayashi S, Sakata Y, Tanaka A, Toda K, Noda T, Nochioka K, Hatano M, Hidaka T, Fujino T, Makita S, Yamaguchi O, Ikeda U, Kimura T, Kohsaka S, Kosuge M, Yamagishi M, Yamashina A. JCS 2017/JHFS 2017 Guideline on Diagnosis and Treatment of Acute and Chronic Heart Failure - Digest Version. Circ J 2019; 83:2084-2184. [PMID: 31511439 DOI: 10.1253/circj.cj-19-0342] [Citation(s) in RCA: 487] [Impact Index Per Article: 81.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Affiliation(s)
- Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences
| | | | - Hiroshi Ito
- Department of Cardiovascular and Respiratory Medicine, Akita University Graduate School of Medicine
| | - Hiroshi Ito
- Department of Cardiovascular Medicine, Division of Biophysiological Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Ken Okumura
- Division of Cardiology, Saiseikai Kumamoto Hospital Cardiovascular Center
| | - Minoru Ono
- Department of Cardiac Surgery, Graduate School of Medicine, The University of Tokyo
| | - Masafumi Kitakaze
- Department of Clinical Medicine and Development, National Cerebral and Cardiovascular Center
| | | | - Yasuki Kihara
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | | | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Yoshikatsu Saiki
- Department of Cardiovascular Surgery, Tohoku University Graduate School of Medicine
| | - Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Naoki Sato
- Department of Cardiovascular Medicine, Kawaguchi Cardiovascular and Respiratory Hospital
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine
| | - Akira Shiose
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | | | - Koichi Node
- Department of Cardiovascular Medicine, Saga University
| | - Taiki Higo
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences
| | - Atsushi Hirayama
- The Division of Cardiology, Department of Medicine, Nihon University Graduate School of Medicine
| | | | - Tohru Masuyama
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | | | - Masafumi Yano
- Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine
| | - Kenji Yamazaki
- Department of Cardiology Surgery, Tokyo Women's Medical University
| | - Kazuhiro Yamamoto
- Department of Molecular Medicine and Therapeutics, Faculty of Medicine, Tottori University
| | | | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Masatoshi Akiyama
- Department of Cardiovascular Surgery, Tohoku University Graduate School of Medicine
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine
| | - Shiro Ishihara
- Department of Cardiology, Nippon Medical School Musashi-Kosugi Hospital
| | - Takayuki Inomata
- Department of Cardiovascular Medicine, Kitasato University Kitasato Institute Hospital
| | | | - Yu-Ki Iwasaki
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Tomohito Ohtani
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | | | - Takatoshi Kasai
- Cardiovascular Respiratory Sleep Medicine, Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine
| | - Mahoto Kato
- Department of Cardiovascular Medicine, Nihon University Graduate School of Medicine
| | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | | | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine
| | - Toru Kuratani
- Department of Minimally Invasive Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Shigeki Kobayashi
- Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine
| | - Yasuhiko Sakata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | | | - Koichi Toda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine
| | - Takashi Noda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Kotaro Nochioka
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| | - Masaru Hatano
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | | | - Takeo Fujino
- Department of Advanced Cardiopulmonary Failure, Kyushu University Graduate School of Medical Sciences
| | - Shigeru Makita
- Department of Cardiac Rehabilitation, Saitama Medical University International Medical Center
| | - Osamu Yamaguchi
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | | | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine and Faculty of Medicine, Kyoto University
| | - Shun Kohsaka
- Department of Cardiology, Keio University School of Medicine
| | - Masami Kosuge
- Division of Cardiology, Yokohama City University Medical Center
| | - Masakazu Yamagishi
- Department of Cardiovascular and Internal Medicine, Kanazawa University Graduate School of Medicine
| | - Akira Yamashina
- Medical Education Promotion Center, Tokyo Medical University
| |
Collapse
|
27
|
Yamashita K, Morimoto S, Seki Y, Watanabe D, Ichihara A. Serum-soluble (pro)renin receptor concentration as a biomarker for organ damage in primary aldosteronism. Hypertens Res 2019; 42:1951-1960. [PMID: 31409916 DOI: 10.1038/s41440-019-0312-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 06/23/2019] [Accepted: 07/16/2019] [Indexed: 12/28/2022]
Abstract
Primary aldosteronism is characterized by inappropriate overproduction of aldosterone by adrenal lesions and leads to hypertension. Excess aldosterone causes organ damage; therefore, finding a biomarker for organ damage risk is vital. The (pro)renin receptor regulates the tissue renin-angiotensin-aldosterone system. The blood soluble (pro)renin receptor concentration is a candidate biomarker that reflects the activity of the tissue renin-angiotensin-aldosterone system. This study investigated the relationships between serum soluble (pro)renin receptor concentrations and indices of organ damage in patients with primary aldosteronism. We examined plasma aldosterone and serum soluble (pro)renin receptor concentrations in patients with primary aldosteronism and evaluated the relationships between these values and organ damage indices, such as the cardio-ankle vascular index, urinary albumin excretion, estimated glomerular filtration rate, and high-sensitivity C-reactive protein levels. We enrolled 121 patients with primary aldosteronism (46 males, 54.9 ± 12.2 years of age). Serum soluble (pro)renin receptor concentrations were significantly positively correlated with the cardio-ankle vascular index, urinary albumin excretion, and high-sensitivity C-reactive protein levels and negatively associated with estimated glomerular filtration rates, independent of other factors. Plasma aldosterone concentrations showed no significant relationships with these indices. In patients with primary aldosteronism, serum soluble (pro)renin receptor concentrations, but not plasma aldosterone concentrations, showed significant associations with organ damage, suggesting that the serum soluble (pro)renin receptor level could be a high-risk biomarker of organ damage.
Collapse
Affiliation(s)
- Kaoru Yamashita
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Satoshi Morimoto
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan.
| | - Yasufumi Seki
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Daisuke Watanabe
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Atsuhiro Ichihara
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
28
|
Leader CJ, Moharram M, Coffey S, Sammut IA, Wilkins GW, Walker RJ. Myocardial global longitudinal strain: An early indicator of cardiac interstitial fibrosis modified by spironolactone, in a unique hypertensive rat model. PLoS One 2019; 14:e0220837. [PMID: 31404095 PMCID: PMC6690508 DOI: 10.1371/journal.pone.0220837] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/25/2019] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVES Is global longitudinal strain (GLS) a more accurate non-invasive measure of histological myocardial fibrosis than left ventricular ejection fraction (LVEF) in a hypertensive rodent model. BACKGROUND Hypertension results in left ventricular hypertrophy and cardiac dysfunction. Speckle-tracking echocardiography has emerged as a robust technique to evaluate cardiac function in humans compared with standard echocardiography. However, its use in animal studies is less clearly defined. METHODS Cyp1a1Ren2 transgenic rats were randomly assigned to three groups; normotensive, untreated hypertensive or hypertensive with daily administration of spironolactone (human equivalent dose of 50 mg/day). Cardiac function and interstitial fibrosis development were monitored for three months. RESULTS The lower limit of normal LVEF was calculated to be 75%. After three months hypertensive animals (196±21 mmHg systolic blood pressure (SBP)) showed increased cardiac fibrosis (8.8±3.2% compared with 2.4±0.7% % in normals), reduced LVEF (from 81±2% to 67±7%) and impaired myocardial GLS (from -17±2% to -11±2) (all p<0.001). Myocardial GLS demonstrated a stronger correlation with cardiac interstitial fibrosis (r2 = 0.58, p<0.0001) than LVEF (r2 = 0.37, p<0.006). Spironolactone significantly blunted SBP elevation (184±15, p<0.01), slowed the progression of cardiac fibrosis (4.9±1.4%, p<0.001), reduced the decline in LVEF (72±4%, p<0.05) and the degree of impaired myocardial GLS (-13±1%, p<0.01) compared to hypertensive animals. CONCLUSIONS This study has demonstrated that, myocardial GLS is a more accurate non-invasive measure of histological myocardial fibrosis compared to standard echocardiography, in an animal model of both treated and untreated hypertension. Spironolactone blunted the progression of cardiac fibrosis and deterioration of myocardial GLS.
Collapse
Affiliation(s)
| | | | - Sean Coffey
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | - Ivan A. Sammut
- Department of Pharmacology, University of Otago, Dunedin, New Zealand
| | | | - Robert J. Walker
- Department of Medicine, University of Otago, Dunedin, New Zealand
| |
Collapse
|
29
|
Brandt MM, Nguyen ITN, Krebber MM, van de Wouw J, Mokry M, Cramer MJ, Duncker DJ, Verhaar MC, Joles JA, Cheng C. Limited synergy of obesity and hypertension, prevalent risk factors in onset and progression of heart failure with preserved ejection fraction. J Cell Mol Med 2019; 23:6666-6678. [PMID: 31368189 PMCID: PMC6787495 DOI: 10.1111/jcmm.14542] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/11/2019] [Accepted: 06/16/2019] [Indexed: 12/15/2022] Open
Abstract
Obesity and hypertension are prevalent comorbidities in heart failure with preserved ejection fraction. To clarify if and how interaction between these comorbidities contributes to development of diastolic dysfunction, lean and obese ZSF1 rats were treated with deoxycorticosterone acetate implants and a high‐salt diet (DS) to induce severe hypertension, or with placebo. In addition to echocardiographic, metabolic and hemodynamic analyses, immunohistochemistry and RNAseq were performed on left ventricular tissue. Obesity negatively affected cardiac output, led to an elevated E/e’ ratio and mildly reduced ejection fraction. DS‐induced hypertension did not affect cardiac output and minimally elevated E/e’ ratio. Diastolic derangements in placebo‐treated obese rats developed in absence of inflammation and fibrosis, yet in presence of oxidative stress and hypertrophic remodelling. In contrast, hypertension triggered apoptosis, inflammation and fibrosis, with limited synergy of the comorbidities observed for inflammation and fibrosis. Transcriptional data suggested that these comorbidities exerted opposite effects on mitochondrial function. In placebo‐treated obese rats, genes involved in fatty acid metabolism were up‐regulated, whereas DS‐induced a down‐regulation of genes involved in oxidative phosphorylation. Overall, limited interaction was observed between these comorbidities in development of diastolic dysfunction. Importantly, differences in obesity‐ and hypertension‐induced cardiac remodelling emphasize the necessity for comorbidity‐specific phenotypical characterization.
Collapse
Affiliation(s)
- Maarten M Brandt
- Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Isabel T N Nguyen
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Merle M Krebber
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jens van de Wouw
- Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Michal Mokry
- Epigenomics facility, University Medical Center Utrecht, Utrecht, The Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maarten J Cramer
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dirk J Duncker
- Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Cheng
- Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
30
|
Butts B, Calhoun DA, Denney TS, Lloyd SG, Gupta H, Gaddam KK, Aban I, Oparil S, Sanders PW, Patel R, Collawn JF, Dell'Italia LJ. Plasma xanthine oxidase activity is related to increased sodium and left ventricular hypertrophy in resistant hypertension. Free Radic Biol Med 2019; 134:343-349. [PMID: 30695690 PMCID: PMC6588431 DOI: 10.1016/j.freeradbiomed.2019.01.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/30/2018] [Accepted: 01/22/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND The extra-renal effects of aldosterone on left ventricular (LV) structure and function are exacerbated by increased dietary sodium in persons with hypertension. Previous studies demonstrated endothelial dysfunction and increased oxidative stress with high salt diet in normotensive salt-resistant subjects. We hypothesized that increased xanthine oxidase (XO), a product of endothelial cells, is related to 24-h urinary sodium and to LV hypertrophy and function in patients with resistant hypertension (RHTN). METHODS The study group included persons with RHTN (n = 91), defined as a blood pressure > 140/90 mmHg on ≥ 3 medications at pharmacologically effective doses. Plasma XO activity and 24-h urine were collected, and cardiac magnetic resonance imaging (MRI) was performed to assess LV function and morphology. Sixty-seven normotensive persons on no cardiovascular medications served as controls. A subset of RHTN (n = 19) received spironolactone without salt restriction for six months with follow-up XO activity measurements and MRI analyses. RESULTS XO activity was increased two-fold in RHTN vs. normal and was positively correlated with LV mass, LV diastolic function, and 24-h urinary sodium. In RHTN patients receiving spironolactone without salt restriction, LV mass decreased, but LV diastolic function and XO activity did not improve. Baseline urinary sodium was positively associated with rate of change of LV mass to volume ratio and the LV E/A ratio. CONCLUSIONS These results demonstrate a potential role of endothelium-derived oxidative stress and excess dietary salt in the pathophysiology of LV hypertrophy and diastolic dysfunction in persons with RHTN unaffected by the addition of spironolactone.
Collapse
Affiliation(s)
- Brittany Butts
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA; Nell Hodgson Woodruff School of Nursing, Emory University, USA
| | - David A Calhoun
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA
| | - Thomas S Denney
- Department of Electrical and Computer Engineering, Auburn University, USA
| | - Steven G Lloyd
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA
| | - Himanshu Gupta
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA; Birmingham Department of Veterans Affairs Medical Center, USA
| | - Krishna K Gaddam
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA
| | - Inmaculada Aban
- Department of Biostatistics, University of Alabama at Birmingham, USA
| | - Suzanne Oparil
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA
| | - Paul W Sanders
- Division of Nephrology, University of Alabama at Birmingham School of Medicine, USA
| | - Rakesh Patel
- Center for Free Radical Biology and Department of Pathology, University of Alabama at Birmingham, USA
| | - James F Collawn
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, USA
| | - Louis J Dell'Italia
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, USA; Birmingham Department of Veterans Affairs Medical Center, USA.
| |
Collapse
|
31
|
Ames MK, Atkins CE, Pitt B. The renin-angiotensin-aldosterone system and its suppression. J Vet Intern Med 2019; 33:363-382. [PMID: 30806496 PMCID: PMC6430926 DOI: 10.1111/jvim.15454] [Citation(s) in RCA: 268] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic activation of the renin-angiotensin-aldosterone system (RAAS) promotes and perpetuates the syndromes of congestive heart failure, systemic hypertension, and chronic kidney disease. Excessive circulating and tissue angiotensin II (AngII) and aldosterone levels lead to a pro-fibrotic, -inflammatory, and -hypertrophic milieu that causes remodeling and dysfunction in cardiovascular and renal tissues. Understanding of the role of the RAAS in this abnormal pathologic remodeling has grown over the past few decades and numerous medical therapies aimed at suppressing the RAAS have been developed. Despite this, morbidity from these diseases remains high. Continued investigation into the complexities of the RAAS should help clinicians modulate (suppress or enhance) components of this system and improve quality of life and survival. This review focuses on updates in our understanding of the RAAS and the pathophysiology of AngII and aldosterone excess, reviewing what is known about its suppression in cardiovascular and renal diseases, especially in the cat and dog.
Collapse
Affiliation(s)
- Marisa K Ames
- Department of Clinical Sciences, College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| | - Clarke E Atkins
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Bertram Pitt
- Department of Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan
| |
Collapse
|
32
|
Young MJ, Adler GK. Aldosterone, the Mineralocorticoid Receptor and Mechanisms of Cardiovascular Disease. VITAMINS AND HORMONES 2019; 109:361-385. [DOI: 10.1016/bs.vh.2018.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
33
|
Mihailidou AS, Tzakos AG, Ashton AW. Non-Genomic Effects of Aldosterone. VITAMINS AND HORMONES 2019; 109:133-149. [DOI: 10.1016/bs.vh.2018.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
34
|
Syed M, Ball JP, Mathis KW, Hall ME, Ryan MJ, Rothenberg ME, Yanes Cardozo LL, Romero DG. MicroRNA-21 ablation exacerbates aldosterone-mediated cardiac injury, remodeling, and dysfunction. Am J Physiol Endocrinol Metab 2018; 315:E1154-E1167. [PMID: 30153065 PMCID: PMC6336952 DOI: 10.1152/ajpendo.00155.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/30/2018] [Accepted: 08/24/2018] [Indexed: 12/21/2022]
Abstract
Primary aldosteronism is characterized by excess aldosterone secretion by the adrenal gland independent of the renin-angiotensin system and accounts for ~10% of hypertensive patients. Excess aldosterone causes cardiac hypertrophy, fibrosis, inflammation, and hypertension. The molecular mechanisms that trigger the onset and progression of aldosterone-mediated cardiac injury remain incompletely understood. MicroRNAs (miRNAs) are endogenous, small, noncoding RNAs that have been implicated in multiple cardiac pathologies; however, their regulation and role in aldosterone-mediated cardiac injury and dysfunction remains mostly unknown. We previously reported that microRNA-21 (miR-21) is the most upregulated miRNA by excess aldosterone in the left ventricle in a rat experimental model of primary aldosteronism. To elucidate the role of miR-21 in aldosterone-mediated cardiac injury and dysfunction, miR-21 knockout mice and their wild-type littermates were treated with aldosterone infusion and salt in the drinking water for 2 or 8 wk. miR-21 genetic ablation exacerbated aldosterone/salt-mediated cardiac hypertrophy and cardiomyocyte cross-sectional area. Furthermore, miR-21 genetic ablation increased the cardiac expression of fibrosis and inflammation markers and fetal gene program. miR-21 genetic ablation increased aldosterone/salt-mediated cardiac dysfunction but did not affect aldosterone/salt-mediated hypertension. miR-21 target gene Sprouty 2 may be implicated in the cardiac effects of miR-21 genetic ablation. Our study shows that miR-21 genetic ablation exacerbates aldosterone/salt-mediated cardiac hypertrophy, injury, and dysfunction blood pressure independently. These results suggest that miR-21 plays a protective role in the cardiac pathology triggered by excess aldosterone. Furthermore, miR-21 supplementation may be a novel therapeutic approach to abolish or mitigate excess aldosterone-mediated cardiovascular deleterious effects in primary aldosteronism.
Collapse
Affiliation(s)
- Maryam Syed
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jana P Ball
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Keisa W Mathis
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Michael E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
- Department of Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
- Women's Health Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Cardio Renal Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- G.V. (Sonny) Montgomery Veterans Affairs Medical Center , Jackson, Mississippi
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Licy L Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Medicine, University of Mississippi Medical Center , Jackson, Mississippi
- Women's Health Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Cardio Renal Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center , Jackson, Mississippi
| | - Damian G Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
- Women's Health Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Cardio Renal Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
35
|
Rouet‐Benzineb P, Merval R, Polidano E. Effects of hypoestrogenism and/or hyperaldosteronism on myocardial remodeling in female mice. Physiol Rep 2018; 6:e13912. [PMID: 30430766 PMCID: PMC6236131 DOI: 10.14814/phy2.13912] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/12/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022] Open
Abstract
We investigated the potential adverse effects of hyperaldosteronism and/or hypoestrogenism on cardiac phenotype, and examined their combined effects in female mice overexpressing cardiac aldosterone synthase (AS). We focused on some signaling cascades challenging defensive responses to adapt and/or to survive in the face of double deleterious stresses, such as Ca2+ -homeostasis, pro/anti-hypertrophic, endoplasmic reticulum stress (ER stress), pro- or anti-apoptotic effectors, and MAP kinase activation, and redox signaling. These protein expressions were assessed by immunoblotting at 9 weeks after surgery. Female wild type (FWT) and FAS mice were fed with phytoestrogen-free diet; underwent ovariectomy (Ovx) or sham-operation (Sham). Ovx increased gain weight and hypertrophy index. Transthoracic echocardiograghy was performed. Both Ovx-induced heart rate decrease and fractional shortening increase were associated with collagen type III shift. Cardiac estrogen receptor (ERα, ERβ) protein expression levels were downregulated in Ovx mice. Hypoestrogenism increased plasma aldosterone and MR protein expression in FAS mice. Both aldosterone and Ovx played as mirror effects on up and downstream signaling effectors of calcium/redox homeostasis, apoptosis, such as concomitant CaMKII activation and calcineurin down-regulation, MAP kinase inhibition (ERK1/2, p38 MAPK) and Akt activation. The ratio Bcl2/Bax is in favor to promote cell survivor. Finally, myocardium had dynamically orchestrated multiple signaling cascades to restore tolerance to hostile environment thereby contributing to a better maintenance of Ca2+ /redox homeostasis. Ovx-induced collagen type III isoform shift and its upregulation may be important for the biomechanical transduction of the heart and the recovery of cardiac function in FAS mice. OVX antagonized aldosterone signaling pathways.
Collapse
|
36
|
Grytaas MA, Sellevåg K, Thordarson HB, Husebye ES, Løvås K, Larsen TH. Cardiac magnetic resonance imaging of myocardial mass and fibrosis in primary aldosteronism. Endocr Connect 2018; 7:413-424. [PMID: 29440130 PMCID: PMC5834771 DOI: 10.1530/ec-18-0039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 02/12/2018] [Indexed: 01/24/2023]
Abstract
BACKGROUND Primary aldosteronism (PA) is associated with increased cardiovascular morbidity, presumably due to left ventricular (LV) hypertrophy and fibrosis. However, the degree of fibrosis has not been extensively studied. Cardiac magnetic resonance imaging (CMR) contrast enhancement and novel sensitive T1 mapping to estimate increased extracellular volume (ECV) are available to measure the extent of fibrosis. OBJECTIVES To assess LV mass and fibrosis before and after treatment of PA using CMR with contrast enhancement and T1 mapping. METHODS Fifteen patients with newly diagnosed PA (PA1) and 24 age- and sex-matched healthy subjects (HS) were studied by CMR with contrast enhancement. Repeated imaging with a new scanner with T1 mapping was performed in 14 of the PA1 and 20 of the HS median 18 months after specific PA treatment and in additional 16 newly diagnosed PA patients (PA2). RESULTS PA1 had higher baseline LV mass index than HS (69 (53-91) vs 51 (40-72) g/m2; P < 0.001), which decreased significantly after treatment (58 (40-86) g/m2; P < 0.001 vs baseline), more with adrenalectomy (n = 8; -9 g/m2; P = 0.003) than with medical treatment (n = 6; -5 g/m2; P = 0.075). No baseline difference was found in contrast enhancement between PA1 and HS. T1 mapping showed no increase in ECV as a myocardial fibrosis marker in PA. Moreover, ECV was lower in the untreated PA2 than HS 10 min post-contrast, and in both PA groups compared with HS 20 min post-contrast. CONCLUSION Specific treatment rapidly reduced LV mass in PA. Increased myocardial fibrosis was not found and may not represent a common clinical problem.
Collapse
Affiliation(s)
- Marianne Aa Grytaas
- Department of Clinical ScienceUniversity of Bergen, Bergen, Norway
- Department of MedicineHaukeland University Hospital, Bergen, Norway
| | - Kjersti Sellevåg
- Department of Heart DiseaseHaukeland University Hospital, Bergen, Norway
| | - Hrafnkell B Thordarson
- Department of Clinical ScienceUniversity of Bergen, Bergen, Norway
- Department of MedicineHaukeland University Hospital, Bergen, Norway
| | - Eystein S Husebye
- Department of Clinical ScienceUniversity of Bergen, Bergen, Norway
- Department of MedicineHaukeland University Hospital, Bergen, Norway
| | - Kristian Løvås
- Department of Clinical ScienceUniversity of Bergen, Bergen, Norway
- Department of MedicineHaukeland University Hospital, Bergen, Norway
| | - Terje H Larsen
- Department of Heart DiseaseHaukeland University Hospital, Bergen, Norway
- Department of BiomedicineUniversity of Bergen, Bergen, Norway
| |
Collapse
|
37
|
Valero-Muñoz M, Backman W, Sam F. Murine Models of Heart Failure with Preserved Ejection Fraction: a "Fishing Expedition". JACC Basic Transl Sci 2017; 2:770-789. [PMID: 29333506 PMCID: PMC5764178 DOI: 10.1016/j.jacbts.2017.07.013] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 12/28/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by signs and symptoms of HF in the presence of a normal left ventricular (LV) ejection fraction (EF). Despite accounting for up to 50% of all clinical presentations of HF, the mechanisms implicated in HFpEF are poorly understood, thus precluding effective therapy. The pathophysiological heterogeneity in the HFpEF phenotype also contributes to this disease and likely to the absence of evidence-based therapies. Limited access to human samples and imperfect animal models that completely recapitulate the human HFpEF phenotype have impeded our understanding of the mechanistic underpinnings that exist in this disease. Aging and comorbidities such as atrial fibrillation, hypertension, diabetes and obesity, pulmonary hypertension and renal dysfunction are highly associated with HFpEF. Yet, the relationship and contribution between them remains ill-defined. This review discusses some of the distinctive clinical features of HFpEF in association with these comorbidities and highlights the advantages and disadvantage of commonly used murine models, used to study the HFpEF phenotype.
Collapse
Affiliation(s)
- Maria Valero-Muñoz
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Warren Backman
- Evans Department of Internal Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Flora Sam
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
- Evans Department of Internal Medicine, Boston University School of Medicine, Boston, Massachusetts
- Cardiovascular Section, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
38
|
Pemayun TGD, Naibaho RM, Sungkar MA. Biventricular Cardiac Hypertrophy in a Patient with Primary Aldosteronism and Atrial Septal Defect. AMERICAN JOURNAL OF CASE REPORTS 2017; 18:963-971. [PMID: 28878203 PMCID: PMC5601392 DOI: 10.12659/ajcr.902271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Patient: Female, 33 Final Diagnosis: Primary aldosteronism • heart failure • atrial septal defect • biventricular cardiac hypertrophy Symptoms: Dyspneu • muscular weakness Medication: Spironolactone • ACE inhibitor Clinical Procedure: Captopril suppression test • adrenalectomy • right cardiac catheterization Specialty: Endocrinology and Metabolism • Cardiology
Collapse
Affiliation(s)
- Tjokorda Gde Dalem Pemayun
- Subdivision of Endocrinology, Metabolism and Diabetes, Department of Medicine, Medical Faculty of Diponegoro University and Dr. Kariadi General Hospital, Semarang, Indonesia
| | - Ridho M Naibaho
- Department of Medicine, Medical Faculty of Diponegoro University and Dr. Kariadi General Hospital, Semarang, Indonesia
| | - Muhammad Achmad Sungkar
- Subdivision of Cardiovascular Medicine, Department of Medicine, Medical Faculty of Diponegoro University and Dr. Kariadi General Hospital, Semarang, Indonesia
| |
Collapse
|
39
|
Fletcher EK, Morgan J, Kennaway DR, Bienvenu LA, Rickard AJ, Delbridge LMD, Fuller PJ, Clyne CD, Young MJ. Deoxycorticosterone/Salt-Mediated Cardiac Inflammation and Fibrosis Are Dependent on Functional CLOCK Signaling in Male Mice. Endocrinology 2017; 158:2906-2917. [PMID: 28911177 DOI: 10.1210/en.2016-1911] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 07/13/2017] [Indexed: 12/19/2022]
Abstract
Activation of the mineralocorticoid receptor (MR) promotes inflammation, fibrosis, and hypertension. Clinical and experimental studies show that MR antagonists have significant therapeutic benefit for all-cause heart failure; however, blockade of renal MRs limits their widespread use. Identification of key downstream signaling mechanisms for the MR in the cardiovascular system may enable development of targeted MR antagonists with selectivity for pathological MR signaling and lower impact on physiological renal electrolyte handling. One candidate pathway is the circadian clock, the dysregulation of which is associated with cardiovascular diseases. We have previously shown that the circadian gene Per2 is dysregulated in hearts with selective deletion of cardiomyocyte MR. We therefore investigated MR-mediated cardiac inflammation and fibrosis in mice that lack normal regulation and oscillation of the circadian clock in peripheral tissues, that is, CLOCKΔ19 mutant mice. The characteristic cardiac inflammatory/fibrotic response to a deoxycorticosterone (DOC)/salt for 8 weeks was significantly blunted in CLOCKΔ19 mice when compared with wild-type mice, despite a modest increase at "baseline" for fibrosis and macrophage number in CLOCKΔ19 mice. In contrast, cardiac hypertrophy in response to DOC/salt was significantly greater in CLOCKΔ19 vs wild-type mice. Markers for renal inflammation and fibrosis were similarly attenuated in the CLOCKΔ19 mice given DOC/salt. Moreover, increased CLOCK expression in H9c2 cardiac cells enhanced MR-mediated transactivation of Per1, suggesting cooperative signaling between these transcription factors. This study demonstrates that the full development of MR-mediated cardiac inflammation and fibrosis is dependent on intact signaling by the circadian protein CLOCK.
Collapse
Affiliation(s)
- Elizabeth K Fletcher
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - James Morgan
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - David R Kennaway
- School of Medicine, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Laura A Bienvenu
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Amanda J Rickard
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Lea M D Delbridge
- Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Peter J Fuller
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Medicine, Monash University, Clayton, Victoria 3168, Australia
| | - Colin D Clyne
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Morag J Young
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Medicine, Monash University, Clayton, Victoria 3168, Australia
- Department of Physiology, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
40
|
Robertson S, Diver LA, Alvarez-Madrazo S, Livie C, Ejaz A, Fraser R, Connell JM, MacKenzie SM, Davies E. Regulation of Corticosteroidogenic Genes by MicroRNAs. Int J Endocrinol 2017; 2017:2021903. [PMID: 28852406 PMCID: PMC5568613 DOI: 10.1155/2017/2021903] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/30/2017] [Accepted: 06/18/2017] [Indexed: 12/14/2022] Open
Abstract
The loss of normal regulation of corticosteroid secretion is important in the development of cardiovascular disease. We previously showed that microRNAs regulate the terminal stages of corticosteroid biosynthesis. Here, we assess microRNA regulation across the whole corticosteroid pathway. Knockdown of microRNA using Dicer1 siRNA in H295R adrenocortical cells increased levels of CYP11A1, CYP21A1, and CYP17A1 mRNA and the secretion of cortisol, corticosterone, 11-deoxycorticosterone, 18-hydroxycorticosterone, and aldosterone. Bioinformatic analysis of genes involved in corticosteroid biosynthesis or metabolism identified many putative microRNA-binding sites, and some were selected for further study. Manipulation of individual microRNA levels demonstrated a direct effect of miR-125a-5p and miR-125b-5p on CYP11B2 and of miR-320a-3p levels on CYP11A1 and CYP17A1 mRNA. Finally, comparison of microRNA expression profiles from human aldosterone-producing adenoma and normal adrenal tissue showed levels of various microRNAs, including miR-125a-5p to be significantly different. This study demonstrates that corticosteroidogenesis is regulated at multiple points by several microRNAs and that certain of these microRNAs are differentially expressed in tumorous adrenal tissue, which may contribute to dysregulation of corticosteroid secretion. These findings provide new insights into the regulation of corticosteroid production and have implications for understanding the pathology of disease states where abnormal hormone secretion is a feature.
Collapse
Affiliation(s)
- Stacy Robertson
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Louise A. Diver
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | | | - Craig Livie
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Ayesha Ejaz
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Robert Fraser
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - John M. Connell
- Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Scott M. MacKenzie
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Eleanor Davies
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| |
Collapse
|
41
|
Yin C, Gu W, Gao Y, Li Z, Chen X, Li Z, Wen S. Association of the -344T/C polymorphism in aldosterone synthase gene promoter with left ventricular structure in Chinese Han: A meta-analysis. Clin Exp Hypertens 2017; 39:562-569. [PMID: 28692307 DOI: 10.1080/10641963.2017.1291660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
No consensus view has been published on the relationship between the aldosterone synthase gene (CYP11B2) -344C/T polymorphism and left ventricular hypertrophy (LVH) in Chinese Han. We undertook a meta-analysis to investigate the potential association of this polymorphism and left ventricular structure-related phenotypes, including left ventricular mass (LVM), left ventricular mass index (LVMI), left ventricular end systolic diameter (LVESD), left ventricular end diastolic dimension (LVEDD), left ventricular posterior wall thickness (LVPWT), and interventricular septal wall thickness (IVS). Studies in English and Chinese were found based on a systematic search of Medline, Embase, CNKI, and Wanfang databases. The dominant model (TT vs. TC+CC) and homozygote model (TT vs. CC) were selected to examine the association between the -344C/T polymorphism and LVH. The random-effects model was used to pool data. From a total of 3104 participants, despite the investigation of six echocardiographic indicators, we found no significant association between the -344C/T variant and LVH in the whole group and the subgroup analyses by blood pressure. However, in the subgroup of northern Han Chinese, TT genotype had higher LVPWT than CC genotype and TC genotype (pheterogeneity = 0.4, pvalue = 0.04, 95% CI 0.09 (0.00, 0.18)). In addition, no evidence of publication bias was observed. In conclusion, our meta-analysis indicated that subjects with TT genotype might have higher risk of developing LVH in northern Han Chinese.
Collapse
Affiliation(s)
- Chengqian Yin
- a Department of Cardiology , Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing , China
| | - Wei Gu
- a Department of Cardiology , Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing , China
| | - Yun Gao
- a Department of Cardiology , Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing , China
| | - Zhao Li
- a Department of Cardiology , Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing , China
| | - Xuanzu Chen
- a Department of Cardiology , Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing , China
| | - Zhizhong Li
- a Department of Cardiology , Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing , China
| | - Shaojun Wen
- b Department of Hypertension Research , Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing , China
| |
Collapse
|
42
|
Ruhs S, Nolze A, Hübschmann R, Grossmann C. 30 YEARS OF THE MINERALOCORTICOID RECEPTOR: Nongenomic effects via the mineralocorticoid receptor. J Endocrinol 2017; 234:T107-T124. [PMID: 28348113 DOI: 10.1530/joe-16-0659] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 12/12/2022]
Abstract
The mineralocorticoid receptor (MR) belongs to the steroid hormone receptor family and classically functions as a ligand-dependent transcription factor. It is involved in water-electrolyte homeostasis and blood pressure regulation but independent from these effects also furthers inflammation, fibrosis, hypertrophy and remodeling in cardiovascular tissues. Next to genomic effects, aldosterone elicits very rapid actions within minutes that do not require transcription or translation and that occur not only in classical MR epithelial target organs like kidney and colon but also in nonepithelial tissues like heart, vasculature and adipose tissue. Most of these effects can be mediated by classical MR and its crosstalk with different signaling cascades. Near the plasma membrane, the MR seems to be associated with caveolin and striatin as well as with receptor tyrosine kinases like EGFR, PDGFR and IGF1R and G protein-coupled receptors like AT1 and GPER1, which then mediate nongenomic aldosterone effects. GPER1 has also been named a putative novel MR. There is a close interaction and functional synergism between the genomic and the nongenomic signaling so that nongenomic signaling can lead to long-term effects and support genomic actions. Therefore, understanding nongenomic aldosterone/MR effects is of potential relevance for modulating genomic aldosterone effects and may provide additional targets for intervention.
Collapse
Affiliation(s)
- Stefanie Ruhs
- Julius Bernstein Institute of PhysiologyMartin Luther University Halle-Wittenberg, Halle, Germany
| | - Alexander Nolze
- Julius Bernstein Institute of PhysiologyMartin Luther University Halle-Wittenberg, Halle, Germany
| | - Ralf Hübschmann
- Julius Bernstein Institute of PhysiologyMartin Luther University Halle-Wittenberg, Halle, Germany
| | - Claudia Grossmann
- Julius Bernstein Institute of PhysiologyMartin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
43
|
Abstract
Myocardial injury, mechanical stress, neurohormonal activation, inflammation, and/or aging all lead to cardiac remodeling, which is responsible for cardiac dysfunction and arrhythmogenesis. Of the key histological components of cardiac remodeling, fibrosis either in the form of interstitial, patchy, or dense scars, constitutes a key histological substrate of arrhythmias. Here we discuss current research findings focusing on the role of fibrosis, in arrhythmogenesis. Numerous studies have convincingly shown that patchy or interstitial fibrosis interferes with myocardial electrophysiology by slowing down action potential propagation, initiating reentry, promoting after-depolarizations, and increasing ectopic automaticity. Meanwhile, there has been increasing appreciation of direct involvement of myofibroblasts, the activated form of fibroblasts, in arrhythmogenesis. Myofibroblasts undergo phenotypic changes with expression of gap-junctions and ion channels thereby forming direct electrical coupling with cardiomyocytes, which potentially results in profound disturbances of electrophysiology. There is strong evidence that systemic and regional inflammatory processes contribute to fibrogenesis (i.e., structural remodeling) and dysfunction of ion channels and Ca2+ homeostasis (i.e., electrical remodeling). Recognizing the pivotal role of fibrosis in the arrhythmogenesis has promoted clinical research on characterizing fibrosis by means of cardiac imaging or fibrosis biomarkers for clinical stratification of patients at higher risk of lethal arrhythmia, as well as preclinical research on the development of antifibrotic therapies. At the end of this review, we discuss remaining key questions in this area and propose new research approaches. © 2017 American Physiological Society. Compr Physiol 7:1009-1049, 2017.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Xiao-Ming Gao
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
44
|
Ball JP, Syed M, Marañon RO, Hall ME, KC R, Reckelhoff JF, Yanes Cardozo LL, Romero DG. Role and Regulation of MicroRNAs in Aldosterone-Mediated Cardiac Injury and Dysfunction in Male Rats. Endocrinology 2017; 158:1859-1874. [PMID: 28368454 PMCID: PMC5460923 DOI: 10.1210/en.2016-1707] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/15/2017] [Indexed: 12/21/2022]
Abstract
Primary aldosteronism is characterized by excess aldosterone (ALDO) secretion independent of the renin-angiotensin system and accounts for approximately 10% of hypertension cases. Excess ALDO that is inappropriate for salt intake status causes cardiac hypertrophy, inflammation, fibrosis, and hypertension. The molecular mechanisms that trigger the onset and progression of ALDO-mediated cardiac injury are poorly understood. MicroRNAs (miRNAs) are endogenous, small, noncoding RNAs that have been implicated in diverse cardiac abnormalities, yet very little is known about their regulation and role in ALDO-mediated cardiac injury. To elucidate the regulation of miRNAs in ALDO-mediated cardiac injury, we performed a time-series analysis of left ventricle (LV) miRNA expression. Uninephrectomized male Sprague-Dawley rats were treated with ALDO (0.75 µg/h) infusion and SALT (1.0% NaCl/0.3% KCl) in the drinking water for up to 8 weeks. ALDO/SALT time dependently modulated the expression of multiple miRNAs in the LV. miR-21 was the most upregulated miRNA after 2 weeks of treatment and remained elevated until the end of the study. To elucidate the role of miR-21 in ALDO/SALT-mediated cardiac injury, miR-21 was downregulated by using antagomirs in ALDO/SALT-treated rats. miR-21 downregulation exacerbated ALDO/SALT-mediated cardiac hypertrophy, expression of fibrosis marker genes, interstitial and perivascular fibrosis, OH-proline content, and cardiac dysfunction. These results suggest that ALDO/SALT-mediated cardiac miR-21 upregulation may be a compensatory mechanism that mitigates ALDO/SALT-mediated cardiac deleterious effects. We speculate that miR-21 supplementation would have beneficial effects in reverting or mitigating cardiac injury and dysfunction in patients with primary aldosteronism.
Collapse
Affiliation(s)
- Jana P. Ball
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Maryam Syed
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Rodrigo O. Marañon
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Cardio-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Michael E. Hall
- Cardio-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Roshan KC
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Jane F. Reckelhoff
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Cardio-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Licy L. Yanes Cardozo
- Cardio-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Damian G. Romero
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Cardio-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
| |
Collapse
|
45
|
Ames MK, Atkins CE, Eriksson A, Hess AM. Aldosterone breakthrough in dogs with naturally occurring myxomatous mitral valve disease. J Vet Cardiol 2017; 19:218-227. [PMID: 28576479 DOI: 10.1016/j.jvc.2017.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 03/18/2017] [Accepted: 03/27/2017] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Aldosterone breakthrough (ABT) is the condition in which angiotensin converting enzyme inhibitors (ACEIs) and/or angiotensin receptor blockers fail to effectively suppress the activity of the renin angiotensin aldosterone system. The objective of this study was to determine if ABT occurs in dogs with naturally occurring myxomatous mitral valve disease receiving an ACEI, using the urine aldosterone to creatinine ratio (UAldo:C) as a measure of renin angiotensin aldosterone system activation. ANIMALS, MATERIALS AND METHODS This study includes 39 dogs with myxomatous mitral valve disease. A UAldo:C cut-off definition (derived from a normal population of healthy, adult, and client-owned dogs) was used to determine the prevalence of ABT in this population. Spearman analysis and univariate logistic regression were used to evaluate the relationship between UAldo:C and ABT (yes/no) and eight variables (age, serum K+ concentration, serum creatinine concentration, ACEI therapy duration and ACEI dosage, furosemide therapy duration and furosemide dosage, and urine sample storage time). Finally, the UAldo:C in dogs receiving spironolactone, as part congestive heart failure (CHF) therapy, was compared to dogs with CHF that were not receiving spironolactone. RESULTS The prevalence of ABT was 32% in dogs with CHF and 30% in dogs without CHF. There was no relationship between either the UAldo:C or the likelihood of ABT and the eight variables. Therapy with spironolactone lead to a significant elevation of the UAldo:C. DISCUSSION Using the UAldo:C and a relatively stringent definition of ABT, it appears that incomplete RAAS blockade is common in dogs with MMVD receiving an ACEI. The prevalence of ABT in this canine population mirrors that reported in humans. While the mechanism of ABT is likely multifactorial and still poorly understood, the proven existence of ABT in dogs offers the potential to improve the prognosis for MMVD with the addition of a mineralocorticoid receptor blocker to current therapeutic regimens. CONCLUSIONS Approximately 30% of dogs being treated for heart disease and CHF satisfied the definition of ABT. Identifying patient subpopulations experiencing ABT may help guide future study design and clinical decision-making.
Collapse
Affiliation(s)
- M K Ames
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Dr. Raleigh, NC, 27607, USA.
| | - C E Atkins
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Dr. Raleigh, NC, 27607, USA
| | - A Eriksson
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Dr. Raleigh, NC, 27607, USA
| | - A M Hess
- Department of Statistics, Colorado State University, Ft. Collins, CO, USA
| |
Collapse
|
46
|
Wang EB, Chaudhary A, Waterhouse TH, Dickinson GL. Population Pharmacokinetics of LY2623091 in Patients With Hypertension and Chronic Kidney Disease. J Clin Pharmacol 2017; 57:739-746. [DOI: 10.1002/jcph.865] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 12/02/2016] [Indexed: 11/11/2022]
|
47
|
Reil JC, Tauchnitz M, Tian Q, Hohl M, Linz D, Oberhofer M, Kaestner L, Reil GH, Thiele H, Steendijk P, Böhm M, Neuberger HR, Lipp P. Hyperaldosteronism induces left atrial systolic and diastolic dysfunction. Am J Physiol Heart Circ Physiol 2016; 311:H1014-H1023. [DOI: 10.1152/ajpheart.00261.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 08/09/2016] [Indexed: 11/22/2022]
Abstract
Patients with hypertension and hyperaldosteronism show an increased risk of stroke compared with patients with essential hypertension. Aim of the study was to assess the effects of aldosterone on left atrial function in rats as a potential contributor to thromboembolism. Osmotic mini-pumps delivering 1.5 μg aldosterone/h were implanted in rats subcutaneously (Aldo, n = 39; controls, n = 38). After 8 wk, left ventricular pressure-volume analysis of isolated working hearts was performed, and left atrial systolic and diastolic function was also assessed by atrial pressure-diameter loops. Moreover, left atrial myocytes were isolated to investigate their global and local Ca2+ handling and contractility. At similar heart rates, pressure-volume analysis of isolated hearts and in vivo hemodynamic measurements revealed neither systolic nor diastolic left ventricular dysfunction in Aldo. In particular, atrial filling pressures and atrial size were not increased in Aldo. Aldo rats showed a significant reduction of atrial late diastolic A wave, atrial active work index, and increased V waves. Consistently, in Aldo rats, sarcomere shortening and the amplitude of electrically evoked global Ca2+ transients were substantially reduced. Sarcoplasmic reticulum-Ca2+ content and fractional Ca2+ release were decreased, substantiated by a reduced sarcoplasmic reticulum calcium ATPase activity, resulting from a reduced CAMKII-evoked phosphorylation of phospholamban. Hyperaldosteronism induced atrial systolic and diastolic dysfunction, while atrial size and left ventricular hemodynamics, including filling pressures, were unaffected in rats. The described model suggests a direct causal link between hyperaldosteronism and decreased atrial contractility and diastolic compliance.
Collapse
Affiliation(s)
- Jan-Christian Reil
- Klinik für Innere Medizin III (Kardiologie, Angiologie, Internistische Intensivmedizin), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
- Klinik für Innere Medizin II, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitäres Herzzentrum Lübeck, Universitätsklinikum Schleswig-Holstein, Lübeck, Germany
| | - Marcus Tauchnitz
- Klinik für Innere Medizin III (Kardiologie, Angiologie, Internistische Intensivmedizin), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | - Qinghai Tian
- Institute for Molecular Cell Biology and Research Centre for Molecular Imaging and Screening, Saarland University, Homburg/Saar, Germany
| | - Mathias Hohl
- Klinik für Innere Medizin III (Kardiologie, Angiologie, Internistische Intensivmedizin), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | - Dominik Linz
- Klinik für Innere Medizin III (Kardiologie, Angiologie, Internistische Intensivmedizin), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | - Martin Oberhofer
- Institute for Molecular Cell Biology and Research Centre for Molecular Imaging and Screening, Saarland University, Homburg/Saar, Germany
| | - Lars Kaestner
- Institute for Molecular Cell Biology and Research Centre for Molecular Imaging and Screening, Saarland University, Homburg/Saar, Germany
| | - Gert-Hinrich Reil
- Klinik für Innere Medizin I, Kardiologie, Klinikum Oldenburg, Oldenburg, Germany
| | - Holger Thiele
- Klinik für Innere Medizin II, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitäres Herzzentrum Lübeck, Universitätsklinikum Schleswig-Holstein, Lübeck, Germany
| | - Paul Steendijk
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands; and
| | - Michael Böhm
- Klinik für Innere Medizin III (Kardiologie, Angiologie, Internistische Intensivmedizin), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | - Hans-Ruprecht Neuberger
- Klinik für Innere Medizin III (Kardiologie, Angiologie, Internistische Intensivmedizin), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
- Klinikum Traunstein, Sektion Rhythmologie, Traunstein, Germany
| | - Peter Lipp
- Institute for Molecular Cell Biology and Research Centre for Molecular Imaging and Screening, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
48
|
Nakagawa H, Somekawa S, Onoue K, Kumazawa T, Ueda T, Seno A, Nakada Y, Nakano T, Matsui M, Soeda T, Okayama S, Kawakami R, Kawata H, Okura H, Saito Y. Salt accelerates aldosterone-induced cardiac remodeling in the absence of guanylyl cyclase-A signaling. Life Sci 2016; 165:9-15. [PMID: 27647418 DOI: 10.1016/j.lfs.2016.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/19/2016] [Accepted: 09/16/2016] [Indexed: 11/27/2022]
Abstract
AIMS Excess sodium causes the development of cardiovascular diseases in conjunction with enhancing renin-angiotensin-aldosterone system (RAAS). Natriuretic peptides are sodium regulators and prevent pathological cardiac alterations by counteracting RAAS. However, it is unknown whether natriuretic peptides inhibit the sodium effect in adverse cardiac alterations. Here, we investigated whether excess salt intake could exacerbate cardiac remodeling in mice with impaired natriuretic peptide signaling. MATERIALS AND METHODS Mice lacking the gene encoding the natriuretic peptide receptor, guanylyl cyclase-A (GC-A), and wild-type mice were administered with either a vehicle substance or a subpressor dose of aldosterone (100ng/kg/min), alongside low salt (0.001% NaCl), normal salt (0.6% NaCl), or high salt diets (6.0% NaCl) for four weeks. Mice were then sacrificed and the hearts were evaluated by histology and RT-PCR. KEY FINDINGS Salt load did not induce cardiac changes in vehicle and aldosterone groups in wild-type mice. On the other hand, cardiac hypertrophy and interstitial fibrosis were significantly exacerbated in a salt dependent manner in GC-A knockout (KO) mice administered aldosterone, and were associated with enhanced gene expression relevant to hypertrophy, fibrosis, and oxidative stress conditions. Of note, excess salt intake increased the expression of Sgk1, serum and glucocorticoid responsive kinase-1, in aldosterone-administered GC-A KO mice. These molecular changes were not observed in wild-type mice. SIGNIFICANCE The results of the present study demonstrate that excess salt intake induced cardiac remodeling in conjunction with aldosterone administration in GC-A KO mice, indicating that GC-A signaling attenuated the deleterious salt effect in aldosterone-induced cardiac remodeling.
Collapse
Affiliation(s)
- Hitoshi Nakagawa
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Satoshi Somekawa
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Kenji Onoue
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Takuya Kumazawa
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Tomoya Ueda
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Ayako Seno
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Yasuki Nakada
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Tomoya Nakano
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Masaru Matsui
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Tunenari Soeda
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Satoshi Okayama
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Rika Kawakami
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Hiroyuki Kawata
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Hiroyuki Okura
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Yoshihiko Saito
- First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan.
| |
Collapse
|
49
|
Nehme J, Mercier N, Labat C, Benetos A, Safar ME, Delcayre C, Lacolley P. Differences Between Cardiac and Arterial Fibrosis and Stiffness in Aldosterone-Salt Rats: Effect of Eplerenone. J Renin Angiotensin Aldosterone Syst 2016; 7:31-9. [PMID: 17083071 DOI: 10.3317/jraas.2006.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Background. Previous experiments have studied separately the development of either cardiac or aortic fibrosis and stiffness in aldosterone (Aldo)-salt hypertensive rats. Our aim was to determine in vivo the effects of Aldo and the Aldo receptor antagonist eplerenone (Epl) on simultaneous changes in cardiac and arterial structure and function and their interactions. Methods and Results. Aldo was administered in uninephrectomised Sprague-Dawley rats receiving a high-salt diet from 8 to 12 weeks of age. Three groups of Aldo-salt rats were treated with 1 to 100 mg/kg-1. d-1 Epl by gavage. Arterial elasticity was measured by elastic modulus (Einc)-wall stress curves using medial cross-sectional area (MCSA). The cardiac and arterial walls were analysed by histomorphometry (elastin and collagen), immunohistochemistry (EIIIA fibronectin, Fn), and Northern blot (collagens I and III). Aldo caused increased systolic blood pressure (SBP), carotid Einc, MCSA, and EIIIA Fn with no change in wall stress or elastin and collagen densities. No difference in collagen mRNA levels was detected between groups. During the same period, cardiac mass and collagen mRNA and protein levels increased markedly in the myocardial tissue. Epl normalised collagen in the myocardium, Eincwall stress curves, MCSA, and EIIIA Fn in Aldo rats. These dose-dependent effects were not accompanied by a consistent reduction in SBP and cardiac mass. Conclusions. In exogenous hyperaldosteronism in the rat, Aldo causes independently myocardial collagen and arterial Fn accumulation, the latter being responsible for increased intrinsic carotid stiffness. Epl prevents both cardiac and arterial effects but does not reduce consistently SBP.
Collapse
|
50
|
Ames MK, Atkins CE, Lantis AC, zum Brunnen J. Evaluation of subacute change in RAAS activity (as indicated by urinary aldosterone:creatinine, after pharmacologic provocation) and the response to ACE inhibition. J Renin Angiotensin Aldosterone Syst 2016; 17:1470320316633897. [PMID: 27009288 PMCID: PMC5843907 DOI: 10.1177/1470320316633897] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/04/2016] [Indexed: 11/18/2022] Open
Abstract
Objective: The objective of this study was to evaluate subacute changes in renin–angiotensin–aldosterone system (RAAS) activity during angiotensin-converting enzyme inhibitor (ACEI) therapy in dogs with experimental RAAS activation. Methods: Analysis of data (urine aldosterone:creatinine ratio (UAldo:C) and serum angiotensin-converting enzyme activity), in 31 healthy dogs with furosemide or amlodipine-activated RAAS that received an ACEI. When furosemide or amlodipine activation of RAAS preceded ACEI administration, incomplete RAAS blockade (IRB) was defined as a UAldo:C greater than (a) the dog’s ‘activated’ baseline value or (b) a population-derived cut-off value (mean + 2 SD (>1.0 μg/g) of pretreatment UAldo:C from our population of research dogs). In studies where RAAS activation occurred concurrently with ACEIs, IRB was defined as (a) a UAldo:C greater than either twofold the dog’s prestimulation baseline value or (b) 1.0 µg/g. Dogs were followed for 7–17 days. Results: Serum angiotensin-converting enzyme activity was measured in 19 dogs and was significantly reduced (P<0.0001) after ACEI administration. The overall incidence of IRB, when RAAS activation preceded ACEI administration, was 33% and 8% for definitions (a) and (b), respectively. The overall incidence of IRB, when ACEIs were concurrent with RAAS activation, was 65% and 61% for definitions (a) and (b), respectively. Conclusion: Increases in UAldo:C, despite ACEI administration, is evidence of IRB in this subacute model of experimental RAAS activation and suppression.
Collapse
Affiliation(s)
- Marisa K Ames
- Department of Clinical Sciences, Colorado State University, USA
| | - Clarke E Atkins
- Department of Clinical Sciences, North Carolina State University, USA
| | | | | |
Collapse
|