1
|
Cotter G, Davison BA, Mann DL, Biegus J, Termaaten JM, Pagnesi M, Freund Y, Voors AA, Ponikowski P, Metra M, Mebazaa A. Acute Heart Failure: Transitioning From Symptom-Based Care to Remission. J Card Fail 2025:S1071-9164(25)00046-6. [PMID: 39952388 DOI: 10.1016/j.cardfail.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 02/17/2025]
Abstract
During the past century, the characteristics of patients with heart failure (HF) and acute HF (AHF) have shifted from patients with severe pump failure due to rheumatic, hypertensive and ischemic heart disease to older and more obese patients with multiple severe comorbidities. The pathophysiology of AHF has shifted, in parallel, from that of advanced, end-stage pump failure caused by severe left ventricular dysfunction to age, obesity and comorbidity-related cardiovascular dysfunction combined with neurohormonal and inflammatory dysregulation or "inflammaging." With the advent of neurohormonal blockers leading to improved outcomes of patients with chronic HF, the focus of AHF therapy has also changed from care directed at early symptom improvement to therapies directed toward longer-term improvements in quality of life and outcomes. Studies conducted in the past 5 years suggest that the beneficial effects seen with the 4 pillars of guideline-directed medical therapy for HF, mostly comprising neurohormonal blockade, can be extended to AHF when these therapies are initiated and rapidly uptitrated during admission and after discharge. A recent pilot study, CORTAHF (Effect of Short-Term Prednisone Therapy on CRP Change in Emergency Department Patients With Acute Heart Failure and Elevated Inflammatory Markers), has suggested that these benefits can be extended by treating patients with AHF and markers of inflammatory activation with anti-inflammatory therapies. Future studies should further examine whether combined anti-inflammatory therapy and neurohormonal blockade can lead to the reversal of disrupted underlying pathophysiology and remission in patients with AHF.
Collapse
Affiliation(s)
- Gad Cotter
- Université Paris Cité, INSERM UMR-S 942 (MASCOT), Paris, France; Heart Initiative, Durham, NC, USA; Momentum Research, Durham, NC, USA.
| | - Beth A Davison
- Université Paris Cité, INSERM UMR-S 942 (MASCOT), Paris, France; Heart Initiative, Durham, NC, USA; Momentum Research, Durham, NC, USA
| | - Douglas L Mann
- Cardiovascular Division, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Jan Biegus
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Jozine M Termaaten
- Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Matteo Pagnesi
- Cardiology, ASST Sedale Civili and Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Yonathan Freund
- Sorbonne Université, Improuvant Emergency Care FHU, Paris, France; Emergency Département and Service Mobile d'Urgence et de Réanimation (SMUR), Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Adriaan A Voors
- Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Marco Metra
- Cardiology, ASST Sedale Civili and Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Alexandre Mebazaa
- Université Paris Cité, INSERM UMR-S 942 (MASCOT), Paris, France; Department of Anesthesiology and Critical Care and Burn Unit, Saint-Louis and Labissière Hospitals, FHU PROMICE, DMU Parabola, APHP Nord, Paris, France
| |
Collapse
|
2
|
Schulze F, Schaible J, Goettel M, Tanaka Y, Hohl K, Schultz A, Jang IJ. Phase 1 studies of the safety, tolerability, pharmacokinetics, and pharmacodynamics of BI 690517 (vicadrostat), a novel aldosterone synthase inhibitor, in healthy male volunteers. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03838-0. [PMID: 39899058 DOI: 10.1007/s00210-025-03838-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE In chronic kidney disease (CKD), raised plasma aldosterone levels are strongly associated with adverse cardiorenal outcomes. Current standard of care may improve outcomes; however, elevated aldosterone levels often persist. We report safety results for BI 690517 (vicadrostat), a potent, selective aldosterone synthase inhibitor under investigation for CKD. METHODS Four phase 1 studies of BI 690517 conducted in healthy European/Chinese/Japanese men: two single rising dose (SRD) and two multiple rising dose (MRD) studies. PRIMARY ENDPOINT proportion of participants with investigator-defined drug-related adverse events (AEs). RESULTS Single and multiple doses of BI 690517 ≤ 80 mg (0.7-80 mg [European SRD]; 3-80 mg [Chinese/Japanese SRD and MRD]) were well tolerated. Proportions of participants with drug-related AEs: European SRD, 8.3% (4/48); Chinese/Japanese SRD, 21.4% (12/56); European MRD, 13.9% (10/72); Japanese MRD, 2.8% (1/36). No serious AEs, deaths, or AEs leading to treatment discontinuation were reported; one AE of severe orthostatic hypotension occurred (European SRD). Plasma exposure to BI 690517 increased dose dependently; median time to maximum concentration was 0.50-1.75 h and mean half-life was 4.4-6.3 h. Exposure was slightly higher in Asians versus Europeans and may relate to lower body weight in Asian participants. A standardized high-fat/high-calorie meal reduced the rate, but not extent, of BI 690517 absorption. Plasma aldosterone concentrations decreased markedly 1-2 h after BI 690517 administration; decreases were more pronounced with increasing BI 690517 doses. CONCLUSION BI 690517 was well tolerated and demonstrated dose-dependent inhibition of aldosterone synthesis. Larger studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Friedrich Schulze
- Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim am Rhein, Germany.
| | - Jennifer Schaible
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Markus Goettel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim am Rhein, Germany
| | - Yuko Tanaka
- Nippon Boehringer Ingelheim Co. Ltd., Tokyo, Japan
| | - Kathrin Hohl
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Armin Schultz
- CRS Clinical Research Services, Mannheim GmbH, Mannheim, Germany
| | - In-Jin Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| |
Collapse
|
3
|
Bansal N, Kathuria D, Babu AM, Dhiman S, Lakhanpal S, Prasad KN, Kumar R, Tyagi Y, Kumar B, Singh MP, Gaidhane AM. A perspective on small molecules targeting the renin-angiotensin-aldosterone system and their utility in cardiovascular diseases: exploring the structural insights for rational drug discovery and development. RSC Med Chem 2025:d4md00720d. [PMID: 39925732 PMCID: PMC11803303 DOI: 10.1039/d4md00720d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/12/2025] [Indexed: 02/11/2025] Open
Abstract
Renin-angiotensin-aldosterone system (RAAS) is crucial in cardiovascular homeostasis. Any disruption in this homeostasis often leads to numerous cardiovascular diseases (CVDs) and non-cardiovascular diseases. Small molecules that show ability toward mechanically modulating RAAS components have been developed to address this problem, thus providing opportunities for innovative drug discovery and development. This review is put forth to provide a comprehensive understanding not only on the signaling mechanisms of RAAS that lead to cardiovascular events but also on the use of small molecules targeting the modulation of RAAS components. Further, the detailed descriptions of the drugs affecting the RAAS and their pharmacodynamics, kinetics, and metabolism profiles are provided. This article also covers the limitations of the present therapeutic armory, followed by their mechanistic insights. A brief discussion is offered on the analysis of the chemical space parameters of the drugs affecting RAAS compared to other cardiovascular and renal categories of medications approved by the US FDA. This review provides structural insights and emphasizes the importance of integrating the current therapeutic regimen with pharmacological tactics to accelerate the development of new therapeutics targeting the RAAS components for improved and efficacious cardiovascular outcomes. Finally, chemical spacing parameters of RAAS modulators are provided, which will help in understanding their peculiarities in modulating the RAAS signaling through structural and functional analyses. Furthermore, this review will assist medicinal chemists working in this field in developing better drug regimens with improved selectivity and efficacy.
Collapse
Affiliation(s)
- Nisha Bansal
- Gramothan Vidyapeeth Home Science Girls PG College Sangaria Rajasthan India
| | - Deepika Kathuria
- University Centre for Research and Development, Chandigarh University Gharuan 140413 Punjab India
| | - Arockia M Babu
- Institute of Pharmaceutical Research, GLA University 17, Km Stone, National Highway #2, Delhi-Mathura Road Mathura India
| | - Sonia Dhiman
- Centre of Research Impact and Outcome, Chitkara University Rajpura-140417 Punjab India
| | - Sorabh Lakhanpal
- Division of Research and Development, Lovely Professional University Phagwara 144411 Punjab India
| | - K Nagendra Prasad
- KKR and KSR Institute of Technology and Sciences Guntur 522017 Andhra Pradesh India
| | - Roshan Kumar
- Graphic Era (Deemed to be University) Clement Town Dehradun-248002 India
| | - Yogita Tyagi
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University Prem Nagar Dehradun 248007 Uttarakhand India
| | - Bhupinder Kumar
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus Srinagar, Garhwal-246174 Uttarakhand India
| | - Mahendra Pratap Singh
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University Chennai India
| | - Abhay M Gaidhane
- Jawaharlal Nehru Medical College, and Global Health Academy, School of Epidemiology and Public Health, Datta Meghe Institute of Higher Education Wardha India
| |
Collapse
|
4
|
Heidari A, Ghorbani M, Hassanzadeh S, Rahmanipour E. A review of the interplay between Takotsubo cardiomyopathy and adrenal insufficiency: Catecholamine surge and glucocorticoid deficiency. Prog Cardiovasc Dis 2024; 87:18-25. [PMID: 39389334 DOI: 10.1016/j.pcad.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Takotsubo Cardiomyopathy (TCM) is a transient heart condition often precipitated by stress and characterized by atypical ventricular ballooning. The interplay between TCM and Adrenal Insufficiency (AI), particularly the influence of catecholamine excess and glucocorticoid deficiency on TCM's pathogenesis in individuals with AI, warrants comprehensive exploration for a better understanding of TCM pathophysiology and establishment of potential therapeutic strategies. METHODS We conducted an extensive literature search via PubMed and Google Scholar, targeting reports on AI, heart failure, and cardiomyopathy, supplemented by forward and backward citation tracing. We analyzed 46 cases from 45 reports, assessing the clinical presentation and outcomes in the context of AI categorization. RESULTS In patients with AI, a glucocorticoid deficit appears to exacerbate the myocardial vulnerability to catecholamine toxicity, precipitating TCM. Most conditions were reversible; however, three pre-1990 cases resulted in irreversible outcomes. CONCLUSIONS The investigation into the AI and TCM intersection highlights the pathogenic significance of catecholamines in the absence of glucocorticoids. The data consolidates the hypothesis that glucocorticoid scarcity exacerbates the cardiac susceptibility to catecholaminergic toxicity, potentially triggering TCM. The study affirms glucocorticoids' cardioprotective roles and elucidates how catecholamine surges contribute to TCM pathogenesis, suggesting strategic clinical management adjustments for AI patients to reduce TCM incidence.
Collapse
Affiliation(s)
- Afshin Heidari
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Health Policy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad Ghorbani
- Health Policy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Hassanzadeh
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Health Policy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Elham Rahmanipour
- Health Policy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Hirsch A, Adolf C, Stüfchen I, Beuschlein F, Brüdgam D, Bidlingmaier M, Reincke M, Quinkler M. NT-proBNP levels in patients with primary hyperaldosteronism and autonomous cortisol cosecretion. Eur J Endocrinol 2024; 191:444-456. [PMID: 39343731 DOI: 10.1093/ejendo/lvae119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/24/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
CONTEXT Patients with primary aldosteronism (PA) have higher cardiac comorbidities including more pronounced left ventricular hypertrophy than patients with essential hypertension. OBJECTIVE Autonomous cortisol cosecretion (ACS) is a common subtype in PA associated with a worse metabolic profile. HYPOTHESIS Autonomous cortisol cosecretion may affect myocardial parameters and result in a worse cardiac outcome compared to patients with PA and without ACS. METHODS Three hundred and sixty-seven patients with PA undergoing 1 mg dexamethasone suppression test (DST) and echocardiography at baseline from 2 centers of the German Conn's Registry were included. Follow-up for up to 3.8 years was available in 192 patients. RESULTS Patients with PA and ACS had higher NT-proBNP levels at baseline compared to patients with PA without ACS (114 vs 75.6 pg/mL, P = .02), but showed no difference in echocardiography values. NT-proBNP levels showed a significant positive correlation (r = 0.141, P = .011) with cortisol levels after DST at baseline. In response to therapy of PA, NT-proBNP levels decreased, but remained significantly higher in patients with ACS compared to patients without ACS. At follow-up, left ventricle end-diastolic dimension (LVEDD) decreased significantly only in patients without ACS. Left atrial diameter (LAD) decreased significantly in patients without ACS and in female patients with ACS but not in male patients. Left ventricular mass index (LVMI) significantly improved in female patients without ACS but remained unchanged in female patients with ACS as well as in male patients at follow-up. CONCLUSIONS In patients with PA, concomitant ACS is associated with a worse cardiac profile and only partial recovery even years after initiation of targeted PA therapy.
Collapse
Affiliation(s)
- Anna Hirsch
- Endocrinology in Charlottenburg, 10627 Berlin, Germany
- Clinical Endocrinology CCM, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Christian Adolf
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, 80336 Munich, Germany
| | - Isabel Stüfchen
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, 80336 Munich, Germany
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, 80336 Munich, Germany
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, UniversitätsSpital Zürich (USZ) und Universität Zürich (UZH), 8091 Zurich, Switzerland
- The LOOP Zurich - Medical Research Center, 8044 Zurich, Switzerland
| | - Denise Brüdgam
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, 80336 Munich, Germany
| | - Martin Bidlingmaier
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, 80336 Munich, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, 80336 Munich, Germany
| | | |
Collapse
|
6
|
Joëls M, Karst H, Tasker JG. The emerging role of rapid corticosteroid actions on excitatory and inhibitory synaptic signaling in the brain. Front Neuroendocrinol 2024; 74:101146. [PMID: 39004314 DOI: 10.1016/j.yfrne.2024.101146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
Over the past two decades, there has been increasing evidence for the importance of rapid-onset actions of corticosteroid hormones in the brain. Here, we highlight the distinct rapid corticosteroid actions that regulate excitatory and inhibitory synaptic transmission in the hypothalamus, the hippocampus, basolateral amygdala, and prefrontal cortex. The receptors that mediate rapid corticosteroid actions are located at or close to the plasma membrane, though many of the receptor characteristics remain unresolved. Rapid-onset corticosteroid effects play a role in fast neuroendocrine feedback as well as in higher brain functions, including increased aggression and anxiety, and impaired memory retrieval. The rapid non-genomic corticosteroid actions precede and complement slow-onset, long-lasting transcriptional actions of the steroids. Both rapid and slow corticosteroid actions appear to be indispensable to adapt to a continuously changing environment, and their imbalance can increase an individual's susceptibility to psychopathology.
Collapse
Affiliation(s)
- Marian Joëls
- University Medical Center Groningen, University of Groningen, the Netherlands; University Medical Center Utrecht, Utrecht University, the Netherlands.
| | - Henk Karst
- University Medical Center Utrecht, Utrecht University, the Netherlands; SILS-CNS. University of Amsterdam, the Netherlands.
| | - Jeffrey G Tasker
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, and Southeast Louisiana Veterans Affairs Healthcare System, New Orleans, USA.
| |
Collapse
|
7
|
Sato R, von Haehling S. Targeting obesity for therapeutic intervention in heart failure patients. Expert Rev Cardiovasc Ther 2024; 22:217-230. [PMID: 38864827 DOI: 10.1080/14779072.2024.2363395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/30/2024] [Indexed: 06/13/2024]
Abstract
INTRODUCTION Heart failure with preserved ejection fraction (HFpEF) is a highly heterogeneous syndrome, making it challenging to improve prognosis with pharmacotherapy. Obesity is one of the leading phenotypes of HFpEF, and its prevalence continues to grow worldwide. Consequently, obesity-targeted interventions have attracted attention as a novel treatment strategy for HFpEF. AREAS COVERED The authors review the association between the pathogenesis of obesity and HFpEF and the potential for obesity-targeted pharmacotherapeutic strategies in HFpEF, together with the latest evidence. The literature search was conducted in PubMed up to April 2024. EXPERT OPINION The STEP HFpEF (Semaglutide Treatment Effect in People with obesity and HFpEF) and SELECT (Semaglutide Effects on Cardiovascular Outcomes in People with Overweight or Obesity) trials recently demonstrated that the glucagon-like peptide 1 analogue, semaglutide, improves various aspects of clinical outcomes in obese HFpEF patients and significantly reduces cardiovascular and heart failure events in non-diabetic obese patients, along with a substantial weight loss. Future clinical trials with other incretin mimetics with more potent weight loss and sub-analyses of the SELECT trial may further emphasize the importance of the obesity phenotype-based approach in the treatment of HFpEF.
Collapse
Affiliation(s)
- Ryosuke Sato
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Lower Saxony, Göttingen, Germany
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Lower Saxony, Göttingen, Germany
| |
Collapse
|
8
|
Gomatam CK, Ingale P, Rodriguez G, Munger S, Pomeranets R, Krishna S, Lowe J, Howard ZM, Rafael-Fortney JA. Cell-type specific effects of mineralocorticoid receptor gene expression suggest intercellular communication regulating fibrosis in skeletal muscle disease. Front Physiol 2024; 15:1322729. [PMID: 38737833 PMCID: PMC11082420 DOI: 10.3389/fphys.2024.1322729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/28/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction: Duchenne muscular dystrophy (DMD) is a fatal striated muscle degenerative disease. DMD is caused by loss of dystrophin protein, which results in sarcolemmal instability and cycles of myofiber degeneration and regeneration. Pathology is exacerbated by overactivation of infiltrating immune cells and fibroblasts, which leads to chronic inflammation and fibrosis. Mineralocorticoid receptors (MR), a type of nuclear steroid hormone receptors, are potential therapeutic targets for DMD. MR antagonists show clinical efficacy on DMD cardiomyopathy and preclinical efficacy on skeletal muscle in DMD models. Methods: We have previously generated myofiber and myeloid MR knockout mouse models to dissect cell-specific functions of MR within dystrophic muscles. Here, we compared skeletal muscle gene expression from both knockouts to further define cell-type specific signaling downstream from MR. Results: Myeloid MR knockout increased proinflammatory and profibrotic signaling, including numerous myofibroblast signature genes. Tenascin C was the most highly upregulated fibrotic gene in myeloid MR-knockout skeletal muscle and is a component of fibrosis in dystrophic skeletal muscle. Surprisingly, lysyl oxidase (Lox), canonically a collagen crosslinker, was increased in both MR knockouts, but did not localize to fibrotic regions of skeletal muscle. Lox localized within myofibers, including only a region of quadriceps muscles. Lysyl oxidase like 1 (Loxl1), another Lox family member, was increased only in myeloid MR knockout muscle and localized specifically to fibrotic regions. Discussion: This study suggests that MR signaling in the dystrophic muscle microenvironment involves communication between contributing cell types and modulates inflammatory and fibrotic pathways in muscle disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jill A. Rafael-Fortney
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
9
|
Bayne S, LeFevre J, Olstinske K, Ravindran S, Munusamy S. Renoprotective Effects of Mineralocorticoid Receptor Antagonists Against Diabetic Kidney Disease. Adv Biol (Weinh) 2024; 8:e2300496. [PMID: 38065929 DOI: 10.1002/adbi.202300496] [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: 09/13/2023] [Revised: 10/18/2023] [Indexed: 03/16/2024]
Abstract
Diabetic kidney disease (DKD) is a growing epidemic worldwide and a leading cause of end-stage kidney disease. Mineralocorticoid receptor (MR) blockade using Finerenone is a recently approved therapeutic approach to slow down the progression of DKD in patients with type 2 diabetes in addition to other therapies such as angiotensin-II converting enzyme inhibitors (ACEIs), angiotensin II receptor blockers (ARBs), sodium-glucose co-transporter 2 (SGLT2) inhibitors, and glucagon-like peptide 1 (GLP-1) analogs. This review elaborates on the pathophysiologic pathways activated by aldosterone (the human mineralocorticoid) in DKD, the pharmacology of three different generations of mineralocorticoid receptor antagonists (MRAs), specifically, spironolactone, eplerenone, and finerenone, and the mechanisms by which these MRAs elicit their protective effects on the kidney under diabetic settings.
Collapse
Affiliation(s)
- Sarah Bayne
- Drake University College of Pharmacy and Health Sciences, Des Moines, IA, 50311, USA
| | - James LeFevre
- Drake University College of Pharmacy and Health Sciences, Des Moines, IA, 50311, USA
| | - Kayla Olstinske
- Drake University College of Pharmacy and Health Sciences, Des Moines, IA, 50311, USA
| | | | - Shankar Munusamy
- Department of Pharmaceutical and Administrative Sciences, Drake University College of Pharmacy and Health Sciences, Des Moines, IA, 50311, USA
| |
Collapse
|
10
|
Correale M, Mazzeo P, Tricarico L, Croella F, Fortunato M, Magnesa M, Amatruda M, Alfieri S, Ferrara S, Ceci V, Dattilo G, Mele M, Iacoviello M, Brunetti ND. Pharmacological Anti-Remodelling Effects of Disease-Modifying Drugs in Heart Failure with Reduced Ejection Fraction. Clin Drug Investig 2022; 42:567-579. [PMID: 35726047 DOI: 10.1007/s40261-022-01166-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 11/25/2022]
Abstract
Cardiac remodelling is an adverse phenomenon linked to heart failure progression and an important contributor to heart failure severity. Cardiac remodelling could represent the real therapeutic goal in the treatment of patients with heart failure with reduced ejection fraction, being potentially reversed through different pharmacotherapies. Currently, there are well-established drugs such as angiotensin-converting enzyme inhibitors/angiotensin II receptor blockers and β-blockers with anti-remodelling effects; recently, angiotensin receptor neprilysin inhibitor effects on inhibiting cardiac remodelling (improving N-terminal pro-B-type natriuretic peptide levels, echocardiographic parameters of reverse cardiac remodelling and right ventricular function in patients with heart failure with reduced ejection fraction) were demonstrated. More recently, hemodynamic consequences of gliflozins, reduced cardiac hydrostatic pressure as a possible cause of ventricular remodelling and hypertrophy were proposed to explain potential anti-remodelling effects of gliflozins. Gliflozins exert their cardioprotective effects by attenuating myofibroblast activity and collagen-mediated remodelling. Another postulated mechanism is represented by the reduction in sympathetic activity, through the reduction in renal afferent nervous activity and the suppression of central reflex mechanisms. Benefits of gliflozins on left ventricular hypertrophy, dilation, and systolic and diastolic function were also described. In this review, we aimed to provide a wide overview on cardiac remodelling with a particular focus on possible anti-remodelling effects of angiotensin receptor neprilysin inhibitors and gliflozins.
Collapse
Affiliation(s)
- Michele Correale
- Cardiothoracic Department, Policlinico Riuniti University Hospital, Viale Pinto 1, 71100, Foggia, Italy.
| | - Pietro Mazzeo
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Lucia Tricarico
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Francesca Croella
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Martino Fortunato
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Michele Magnesa
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Marco Amatruda
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Simona Alfieri
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Salvatore Ferrara
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Vincenzo Ceci
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giuseppe Dattilo
- Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Marco Mele
- Cardiothoracic Department, Policlinico Riuniti University Hospital, Viale Pinto 1, 71100, Foggia, Italy
| | - Massimo Iacoviello
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | | |
Collapse
|
11
|
Yaribeygi H, Maleki M, Majeed M, Jamialahmadi T, Sahebkar A. Renoprotective Roles of Curcumin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1328:531-544. [PMID: 34981504 DOI: 10.1007/978-3-030-73234-9_38] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The use of herb-based therapies is increasing over the past decades. These agents have been reported to provide many beneficial effects in many experimental and clinical studies. Curcumin is one of these agents which has potent pharmacological effects enabling it for the prevent and treatment of many diseases and pathologies such as renal disorders, hyperglycemia, oxidative stress, hypertension, and dyslipidemia. However, the exact molecular mechanisms mediating these renoprotective effects of curcumin are not well established. So, in the current study, we surveyed for possible renoprotective roles of curcumin and concluded how curcumin protects against renal injuries.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Mina Maleki
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran.,Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
12
|
Leader CJ, Wilkins GT, Walker RJ. The effect of spironolactone on cardiac and renal fibrosis following myocardial infarction in established hypertension in the transgenic Cyp1a1Ren2 rat. PLoS One 2021; 16:e0260554. [PMID: 34843581 PMCID: PMC8629264 DOI: 10.1371/journal.pone.0260554] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/11/2021] [Indexed: 11/19/2022] Open
Abstract
AIMS The renin-angiotensin-aldosterone axis plays a key role in mediating cardiac and kidney injury. Mineralocorticoid receptor antagonism has beneficial effects on cardiac dysfunction, but effects are less well quantified in the cardiorenal syndrome. This study investigated cardiac and kidney pathophysiology following permanent surgical ligation to induce myocardial infarction (MI) in hypertensive animals with or without mineralocorticoid receptor antagonism. METHODS Hypertension was induced in adult male Cyp1a1Ren2 rats. Hypertensive animals underwent MI surgery (n = 6), and were then treated daily with spironolactone for 28 days with serial systolic blood pressure measurements, echocardiograms and collection of urine and serum biochemical data. They were compared to hypertensive animals (n = 4), hypertensive animals treated with spironolactone (n = 4), and hypertensive plus MI without spironolactone (n = 6). Cardiac and kidney tissue was examined for histological and immunohistochemical analysis. RESULTS MI superimposed on hypertension resulted in an increase in interstitial cardiac fibrosis (p<0.001), renal cortical interstitial fibrosis (p<0.01) and glomerulosclerosis (p<0.01). Increased fibrosis was accompanied by myofibroblast and macrophage infiltration in the heart and the kidney. Spironolactone post-MI, diminished the progressive fibrosis (p<0.001) and inflammation (myofibroblasts (p<0.05); macrophages (p<0.01)) in both the heart and the kidney, despite persistently elevated SBP (182±19 mmHg). Despite the reduction in inflammation and fibrosis, spironolactone did not modify ejection fraction, proteinuria, or renal function when compared to untreated animals post MI. CONCLUSION This model of progressive cardiorenal dysfunction more closely replicates the clinical setting. Mineralocorticoid receptor blockade at a clinically relevant dose, blunted progression of cardiac and kidney fibrosis with reduction in cardiac and kidney inflammatory myofibroblast and macrophage infiltration. Further studies are underway to investigate the combined actions of angiotensin blockade with mineralocorticoid receptor blockade.
Collapse
Affiliation(s)
- C. J. Leader
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | - G. T. Wilkins
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | - R. J. Walker
- Department of Medicine, University of Otago, Dunedin, New Zealand
| |
Collapse
|
13
|
Dumeny L, Vardeny O, Edelmann F, Pieske B, Duarte JD, Cavallari LH. NR3C2 genotype is associated with response to spironolactone in diastolic heart failure patients from the Aldo-DHF trial. Pharmacotherapy 2021; 41:978-987. [PMID: 34569641 DOI: 10.1002/phar.2626] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/21/2021] [Accepted: 09/06/2021] [Indexed: 01/08/2023]
Abstract
STUDY OBJECTIVE This study aimed to determine if variants in NR3C2, which codes the target protein of spironolactone, or CYP11B2, which is involved in aldosterone synthesis, were associated with spironolactone response, focused on the primary end point of diastolic function (E/e'), in Aldosterone Receptor Blockade in Diastolic Heart Failure (Aldo-DHF) participants. DESIGN Post-hoc genetic analysis. DATA SOURCE Data and samples were derived from the multi-center, randomized, double-blind, placebo-controlled Aldo-DHF trial. PATIENTS Aldo-DHF participants treated with spironolactone (n = 184) or placebo (n = 178) were included. INTERVENTION Participants were genotyped for NR3C2 rs5522, NR3C2 rs2070951 and CYP11B2 rs1799998 via pyrosequencing. MEASUREMENTS In the placebo and spironolactone arms, separate multivariable linear regression analyses were performed for change in E/e' with each single nucleotide polymorphism (SNP), adjusted for age, sex, and baseline E/e'. To discern potential mechanisms of a genotype effect, associated SNPs were further examined for their association with change in blood pressure, circulating procollagen type III N-terminal peptide (PIIINP), and left atrial area. MAIN RESULTS Carriers of the rs5522 G allele in the placebo arm had a greater increase in E/e' over the 12-month course of the trial compared to noncarriers (β = 1.10; 95% confidence interval [CI]: 0.05-2.16; p = 0.04). No corresponding E/e' worsening by rs5522 genotype was observed in the spironolactone arm. None of the other genotypes were associated with change in E/e'. Compared to noncarriers, rs5522 G carriers also had a greater increase in left atrial area with placebo (β = 0.83; 95% CI: 0.17-1.48; p = 0.01) and a greater reduction in diastolic blood pressure with spironolactone (β = -3.56; 95% CI: -6.73 to -0.39; p = 0.03). Serum PIIINP levels were similar across rs5522 genotypes. CONCLUSIONS Our results suggest that spironolactone attenuates progression of diastolic dysfunction associated with the NR3C2 rs5522 G allele. Validation of our findings is needed.
Collapse
Affiliation(s)
- Leanne Dumeny
- Center for Pharmacogenomics and Precision Medicine and Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Genetics and Genomics, Genetics Institute, University of Florida, Gainesville, Florida, USA
| | - Orly Vardeny
- Center for Care Delivery and Outcomes Research, Minneapolis Veteran Affairs Health Care System, Minneapolis, Minnesota, USA
| | - Frank Edelmann
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow Klinikum, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow Klinikum, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Department of Cardiology, German Heart Center Berlin, Berlin, Germany
| | - Julio D Duarte
- Center for Pharmacogenomics and Precision Medicine and Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Genetics and Genomics, Genetics Institute, University of Florida, Gainesville, Florida, USA
| | - Larisa H Cavallari
- Center for Pharmacogenomics and Precision Medicine and Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, USA.,Genetics and Genomics, Genetics Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
14
|
Epstein M, Freundlich M. The intersection of Mineralocorticoid Receptor (MR) activation and the FGF23 - Klotho cascade. A Duopoly that promotes renal and cardiovascular injury. Nephrol Dial Transplant 2021; 37:211-221. [PMID: 34459924 DOI: 10.1093/ndt/gfab254] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Indexed: 12/17/2022] Open
Abstract
The nexus of CKD and cardiovascular disease (CVD) amplifies the morbidity and mortality of CKD, emphasizing the need for defining and establishing therapeutic initiatives to modify and abrogate the progression of CKD and concomitant CV risks. In addition to the traditional CV risk factors, disturbances of mineral metabolism are specific risk factors that contribute to the excessive CV mortality in patients with CKD. These risk factors include dysregulations of circulating factors that modulate phosphate metabolism including fibroblast growth factor 23 (FGF23) and soluble Klotho. Reduced circulating levels and suppressed renal klotho expression may be associated with adverse outcomes in CKD patients. While elevated circulating concentrations or locally produced FGF23 in the strained heart exert pro-hypertrophic mechanisms on the myocardium, Klotho attenuates tissue fibrosis, progression of CKD, cardiomyopathy, endothelial dysfunction, vascular stiffness, and vascular calcification. Mineralocorticoid receptor (MR) activation in non-classical targets, mediated by aldosterone and other ligands, amplifies CVD in CKD. In concert, we detail how the interplay of elevated FGF23, activation of the MR, and concomitant reductions of circulating Klotho in CKD, may potentiate each other's deleterious effects on kidney and the heart, thereby contributing to the initiation and progression of kidney and cardiac functional deterioration, acting through multipronged albeit complementary mechanistic pathways.
Collapse
Affiliation(s)
- Murray Epstein
- Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael Freundlich
- Division of Pediatric Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
15
|
Vodošek Hojs N, Bevc S, Ekart R, Piko N, Petreski T, Hojs R. Mineralocorticoid Receptor Antagonists in Diabetic Kidney Disease. Pharmaceuticals (Basel) 2021; 14:561. [PMID: 34208285 PMCID: PMC8230766 DOI: 10.3390/ph14060561] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus is a global health issue and main cause of chronic kidney disease. Both diseases are also linked through high cardiovascular morbidity and mortality. Diabetic kidney disease (DKD) is present in up to 40% of diabetic patients; therefore, prevention and treatment of DKD are of utmost importance. Much research has been dedicated to the optimization of DKD treatment. In the last few years, mineralocorticoid receptor antagonists (MRA) have experienced a renaissance in this field with the development of non-steroidal MRA. Steroidal MRA have known cardiorenal benefits, but their use is limited by side effects, especially hyperkalemia. Non-steroidal MRA still block the damaging effects of mineralocorticoid receptor overactivation (extracellular fluid volume expansion, inflammation, fibrosis), but with fewer side effects (hormonal, hyperkalemia) than steroidal MRA. This review article summarizes the current knowledge and newer research conducted on MRA in DKD.
Collapse
Affiliation(s)
- Nina Vodošek Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia; (S.B.); (T.P.); (R.H.)
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia; (S.B.); (T.P.); (R.H.)
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia;
| | - Robert Ekart
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia;
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia;
| | - Nejc Piko
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia;
| | - Tadej Petreski
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia; (S.B.); (T.P.); (R.H.)
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia;
| | - Radovan Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia; (S.B.); (T.P.); (R.H.)
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia;
| |
Collapse
|
16
|
Watson K, Kukin A, Wasik AK, Shulenberger CE. Nonsteroidal Mineralocorticoid Receptor Antagonists: Exploring Role in Cardiovascular Disease. J Cardiovasc Pharmacol 2021; 77:685-698. [PMID: 34057158 DOI: 10.1097/fjc.0000000000000990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 01/28/2021] [Indexed: 12/20/2022]
Abstract
ABSTRACT Aldosterone, a mineralocorticoid hormone, plays a role in the pathophysiology of many cardiovascular disease states. Mineralocorticoid receptor antagonists (MRAs) have been shown to improve clinical outcomes in select patient populations. However, use of available steroidal receptor antagonists, eplerenone and spironolactone, is often limited by the risk or development of hyperkalemia. Nonsteroidal MRAs have been designed to overcome this limitation. The nonsteroidal MRAs have been studied in patients with heart failure with reduced ejection fraction, hypertension, and to lower the risk of cardiac and renal outcomes in those with type 2 diabetes and renal disease. In this review, the pharmacology of the MRAs is compared, the data evaluating the use of nonsteroidal MRAs are examined, and the place of this new generation of therapy is discussed. At this time, it seems that there could be a future role for nonsteroidal MRAs to reduce the risk of renal outcomes in high-risk individuals.
Collapse
Affiliation(s)
- Kristin Watson
- Department of Pharmacy Practice and Science, University of Maryland School of Pharmacy, Baltimore, MD
- ATRIUM Cardiology Collaborative, Baltimore, MD
| | - Alina Kukin
- Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Allie K Wasik
- Department of Pharmacy, Northwestern Memorial Hospital Bluhm Cardiovascular Institute, Chicago, IL; and
| | | |
Collapse
|
17
|
Ennis IL, Pérez NG. Cardiac Mineralocorticoid Receptor and the Na +/H + Exchanger: Spilling the Beans. Front Cardiovasc Med 2021; 7:614279. [PMID: 33553262 PMCID: PMC7854694 DOI: 10.3389/fcvm.2020.614279] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/01/2020] [Indexed: 01/08/2023] Open
Abstract
Current evidence reveals that cardiac mineralocorticoid receptor (MR) activation following myocardial stretch plays an important physiological role in adapting developed force to sudden changes in hemodynamic conditions. Its underlying mechanism involves a previously unknown nongenomic effect of the MR that triggers redox-mediated Na+/H+ exchanger (NHE1) activation, intracellular Na+ accumulation, and a consequent increase in Ca2+ transient amplitude through reverse Na+/Ca2+ exchange. However, clinical evidence assigns a detrimental role to MR activation in the pathogenesis of severe cardiac diseases such as congestive heart failure. This mini review is meant to present and briefly discuss some recent discoveries about locally triggered cardiac MR signals with the objective of shedding some light on its physiological but potentially pathological consequences in the heart.
Collapse
Affiliation(s)
- Irene Lucía Ennis
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas de la Plata, Universidad Nacional de La Plata, La Plata, Argentina
| | - Néstor Gustavo Pérez
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas de la Plata, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
18
|
Yaribeygi H, Maleki M, Sathyapalan T, Jamialahmadi T, Sahebkar A. Incretin-based therapies and renin-angiotensin system: Looking for new therapeutic potentials in the diabetic milieu. Life Sci 2020; 256:117916. [PMID: 32534034 DOI: 10.1016/j.lfs.2020.117916] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 05/27/2020] [Accepted: 06/04/2020] [Indexed: 02/08/2023]
Abstract
Incretin-based therapies include pharmacologic agents such as glucagon like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors which exert potent anti-hyperglycemic effects in the diabetic milieu. They are also shown to have extra-pancreatic effects. Renin-angiotensin system is part of the endocrine system which is widely distributed in the body and is closely involved in water and electrolyte homeostasis as well as renal and cardiovascular functions. Hence the renin-angiotensin system is the main target for treating patients with various renal and cardiovascular disorders. There is growing evidence that incretins have modulatory effects on renin-angiotensin system activity; thereby, can be promising therapeutic agents for the management of renal and cardiovascular disorders. But the exact molecular interactions between incretins and renin-angiotensin system are not clearly understood. In this current study, we have reviewed the possible molecular mechanisms by which incretins modulate renin-angiotensin system activity.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Mina Maleki
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull
| | - Tannaz Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
| |
Collapse
|
19
|
Leader CJ, Kelly DJ, Sammut IA, Wilkins GT, Walker RJ. Spironolactone mitigates, but does not reverse, the progression of renal fibrosis in a transgenic hypertensive rat. Physiol Rep 2020; 8:e14448. [PMID: 32441493 PMCID: PMC7243196 DOI: 10.14814/phy2.14448] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/15/2020] [Accepted: 04/24/2020] [Indexed: 01/10/2023] Open
Abstract
Hypertension plays an important role in the development and progression of chronic kidney disease. Studies to date, with mineralocorticoid receptor antagonists (MRA), have demonstrated varying degrees of results in modifying the development of renal fibrosis. This study aimed to investigate whether treatment with a MRA commenced following the establishment of hypertension, a situation more accurately representing the clinical setting, modified the progression of renal fibrosis. Using male Cyp1a1Ren2 rats (n = 28), hypertension was established by addition of 0.167% indole-3-carbinol (w/w) to the rat chow, for 2 weeks prior to treatment. Rats were then divided into normotensive, hypertensive (H), or hypertensive with daily oral spironolactone treatment (H + SP) (human equivalent dose 50 mg/day). Physiological data and tissue were collected after 4 and 12 weeks for analysis. After 4 weeks, spironolactone had no demonstrable effect on systolic blood pressure (SBP), proteinuria, or macrophage infiltration in the renal cortex. However, glomerulosclerosis and renal cortical fibrosis were significantly decreased. Following 12 weeks of spironolactone treatment, SBP was lowered (not back to normotensive levels), proteinuria was reduced, and the progression of glomerulosclerosis and renal cortical fibrosis was significantly blunted. This was associated with a significant reduction in macrophage and myofibroblast infiltration, as well as CTGF and pSMAD2 expression. In summary, in a model of established hypertension, spironolactone significantly blunted the progression of renal fibrosis and glomerulosclerosis, and downregulated the renal inflammatory response, which was associated with reduced proteinuria, despite only a partial reduction in systolic blood pressure. This suggests a blood pressure independent effect of MRA on renal fibrosis.
Collapse
Affiliation(s)
| | - Darren J. Kelly
- Department of MedicineUniversity of MelbourneMelbourneVICAustralia
| | - Ivan A. Sammut
- Department of PharmacologyUniversity of OtagoDunedinNew Zealand
| | | | | |
Collapse
|
20
|
Garg A, Foinquinos A, Jung M, Janssen‐Peters H, Biss S, Bauersachs J, Gupta SK, Thum T. MiRNA
‐181a is a novel regulator of aldosterone–mineralocorticoid receptor‐mediated cardiac remodelling. Eur J Heart Fail 2020; 22:1366-1377. [DOI: 10.1002/ejhf.1813] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/26/2020] [Accepted: 03/16/2020] [Indexed: 12/28/2022] Open
Affiliation(s)
- Ankita Garg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School Hannover Germany
| | - Ariana Foinquinos
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School Hannover Germany
| | - Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School Hannover Germany
| | - Heike Janssen‐Peters
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School Hannover Germany
| | - Sinje Biss
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School Hannover Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology Hannover Medical School Hannover Germany
| | - Shashi Kumar Gupta
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School Hannover Germany
- Council of Scientific and Industrial Research ‐ Central Drug Research Institute Lucknow India
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School Hannover Germany
- REBIRTH Center of Translational Regenerative Medicine, Hannover Medical School Hannover Germany
| |
Collapse
|
21
|
Jorba G, Aguirre-Plans J, Junet V, Segú-Vergés C, Ruiz JL, Pujol A, Fernández-Fuentes N, Mas JM, Oliva B. In-silico simulated prototype-patients using TPMS technology to study a potential adverse effect of sacubitril and valsartan. PLoS One 2020; 15:e0228926. [PMID: 32053711 PMCID: PMC7018085 DOI: 10.1371/journal.pone.0228926] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/26/2020] [Indexed: 12/11/2022] Open
Abstract
Unveiling the mechanism of action of a drug is key to understand the benefits and adverse reactions of a medication in an organism. However, in complex diseases such as heart diseases there is not a unique mechanism of action but a wide range of different responses depending on the patient. Exploring this collection of mechanisms is one of the clues for a future personalized medicine. The Therapeutic Performance Mapping System (TPMS) is a Systems Biology approach that generates multiple models of the mechanism of action of a drug. Each molecular mechanism generated could be associated to particular individuals, here defined as prototype-patients, hence the generation of models using TPMS technology may be used for detecting adverse effects to specific patients. TPMS operates by (1) modelling the responses in humans with an accurate description of a protein network and (2) applying a Multilayer Perceptron-like and sampling strategy to find all plausible solutions. In the present study, TPMS is applied to explore the diversity of mechanisms of action of the drug combination sacubitril/valsartan. We use TPMS to generate a wide range of models explaining the relationship between sacubitril/valsartan and heart failure (the indication), as well as evaluating their association with macular degeneration (a potential adverse effect). Among the models generated, we identify a set of mechanisms of action associated to a better response in terms of heart failure treatment, which could also be associated to macular degeneration development. Finally, a set of 30 potential biomarkers are proposed to identify mechanisms (or prototype-patients) more prone of suffering macular degeneration when presenting good heart failure response. All prototype-patients models generated are completely theoretical and therefore they do not necessarily involve clinical effects in real patients. Data and accession to software are available at http://sbi.upf.edu/data/tpms/.
Collapse
Affiliation(s)
- Guillem Jorba
- Anaxomics Biotech SL, Barcelona, Catalonia, Spain
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Experimental and Health Science, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Joaquim Aguirre-Plans
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Experimental and Health Science, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Valentin Junet
- Anaxomics Biotech SL, Barcelona, Catalonia, Spain
- Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Catalonia, Spain
| | | | | | - Albert Pujol
- Anaxomics Biotech SL, Barcelona, Catalonia, Spain
| | - Narcís Fernández-Fuentes
- Department of Biosciences, U Science Tech, Universitat de Vic-Universitat Central de Catalunya, Vic, Catalonia, Spain
| | | | - Baldo Oliva
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Experimental and Health Science, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| |
Collapse
|
22
|
Huang KH, Chen YH, Lee LC, Tai MC, Chung CH, Chen JT, Liang CM, Chien WC, Chen CL. Relationship between heart failure and central serous chorioretinopathy: A cohort study in Taiwan. J Chin Med Assoc 2019; 82:941-947. [PMID: 31805017 DOI: 10.1097/jcma.0000000000000207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Both central serous chorioretinopathy (CSCR) and heart failure (HF) are disorders with a complex pathogenesis, whereas the two diseases might share similar pathogenesis. This study aimed to evaluate whether patients with HF are exposed to potential risk of CSCR by using the National Health Insurance Research Database (NHIRD). METHODS Data were collected from the NHIRD over a 14-year period. Variables were analyzed with the Pearson chi-square test and Fisher's exact test. The risk factors for disease development were examined by adjusted hazard ratio (aHR). Kaplan-Meier analysis was performed to compare the cumulative incidence of CSCR. RESULTS A total of 24 426 patients with HF were enrolled in the study cohort, and there were 24 426 patients without HF in the control cohort. The incidence rate of CSCR was higher in the study cohort than in the control cohort (aHR = 4.572, p < 0.001). CSCR occurred more commonly in males than in females. The overall incidence of CSCR was 30.07 per 100 000 person-years in the study cohort and 23.06 per 100 000 person-years in the control cohort. Besides, subgroup analysis revealed that no matter in gender or age group, HF patients were in an increased risk of CSCR diagnosis (male/female, aHR = 3.268/7.701; 20-59 years/≥60 years, aHR = 3.405/5.501, p < 0.001). CONCLUSION HF is a significant indicator for CSCR. Patients with HF should stay alert for potential disorder of visual impairment. Further prospective studies to investigate the relationship between HF and CSCR could provide more information.
Collapse
Affiliation(s)
- Ke-Hao Huang
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
- Department of Ophthalmology, Song-Shan Branch of Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yi-Hao Chen
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Lung-Chi Lee
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Ming-Cheng Tai
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chi-Hsiang Chung
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC
- Taiwanese Injury Prevention and Safety Promotion Association, Taipei, Taiwan, ROC
| | - Jiann-Torng Chen
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chang-Min Liang
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Wu-Chien Chien
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Ching-Long Chen
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| |
Collapse
|
23
|
Packer M. Obesity-Associated Heart Failure as a Theoretical Target for Treatment With Mineralocorticoid Receptor Antagonists. JAMA Cardiol 2019; 3:883-887. [PMID: 30046826 DOI: 10.1001/jamacardio.2018.2090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Importance Despite their clinical benefits, mineralocorticoid receptor antagonists are greatly underprescribed by most practitioners who treat patients with chronic heart failure. A novel approach to encouraging the use of these drugs is to enhance awareness about the intimate link between aldosterone and obesity. Observations There is a strong association between abdominal obesity and circulating levels of aldosterone, and markers of abdominal obesity identify patients most likely to benefit from mineralocorticoid receptor antagonism. In a trial of patients with heart failure and a reduced ejection fraction, patients with an increased waist circumference exhibited an approximately 50% reduction in the risk of a primary end point. The magnitude of benefit was more than twice as great in patients with abdominal obesity than in those with a normal waist circumference, and patients with abdominal obesity tolerated treatment better than nonobese patients. Similarly, in a trial of patients with heart failure and a preserved ejection fraction, those who were most likely to have abdominal obesity (identified by their level of natriuretic peptides) were most likely to demonstrate a benefit of treatment with spironolactone, exhibiting an approximately 80% reduction in the risk of a primary end point (based on a small number of events). Conclusions and Relevance Although these analyses are post hoc, their concordance and strong biological foundation suggests that abdominal obesity may identify patients who respond most favorably to mineralocorticoid receptor antagonism. Given the easy availability of its measurement, targeting patients with an increased waist circumference could enhance the adoption of these important drugs for the treatment of chronic heart failure in clinical practice.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas
| |
Collapse
|
24
|
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
|
25
|
Armani A, Infante M, Fabbri A, Caprio M. Comment on "mineralocorticoid antagonism enhances brown adipose tissue function in humans: A randomized placebo-controlled cross-over study". Diabetes Obes Metab 2019; 21:2024-2026. [PMID: 31050122 DOI: 10.1111/dom.13756] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/19/2019] [Accepted: 04/30/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Marco Infante
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, Florida
- Unit of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Andrea Fabbri
- Unit of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Primary aldosteronism (PA) is a common form of hypertension characterized by autonomous aldosterone secretion from one or both adrenal glands. The purpose of this review is to synthesize recent research findings regarding cardiovascular disease risk in PA both pre- and post-targeted therapy. RECENT FINDINGS Previously considered a rare disease, recent prevalence studies demonstrate that PA is actually a very common, yet vastly under-diagnosed, etiology of hypertension. Prior to targeted therapy, PA is associated with substantially higher rates of cardiovascular disease compared with essential hypertension. Surgical adrenalectomy is highly effective in curing or improving hypertension as well as mitigating cardiovascular disease risk in patients with unilateral PA. For the remainder of PA patients, MR antagonists are recommended; however, several recent studies have brought into question their effectiveness in improving cardiovascular outcomes. PA is a common cause of hypertension that leads to disproportionately high rates of cardiovascular disease. Future studies are needed to enhance the clinical approach to both identification and treatment of patients with PA to optimize long-term cardiovascular outcomes.
Collapse
Affiliation(s)
- Gregory L Hundemer
- Division of Nephrology, Ottawa Hospital Research Institute, University of Ottawa, 1967 Riverside Drive, Ottawa, ON, K1H 7W9, Canada.
| |
Collapse
|
27
|
Melis N, Thuillier R, Steichen C, Giraud S, Sauvageon Y, Kaminski J, Pelé T, Badet L, Richer JP, Barrera-Chimal J, Jaisser F, Tauc M, Hauet T. Emerging therapeutic strategies for transplantation-induced acute kidney injury: protecting the organelles and the vascular bed. Expert Opin Ther Targets 2019; 23:495-509. [PMID: 31022355 DOI: 10.1080/14728222.2019.1609451] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Renal ischemia-reperfusion injury (IRI) is a significant clinical challenge faced by clinicians in a broad variety of clinical settings such as perioperative and intensive care. Renal IRI induced acute kidney injury (AKI) is a global public health concern associated with high morbidity, mortality, and health-care costs. Areas covered: This paper focuses on the pathophysiology of transplantation-related AKI and recent findings on cellular stress responses at the intersection of 1. The Unfolded protein response; 2. Mitochondrial dysfunction; 3. The benefits of mineralocorticoid receptor antagonists. Lastly, perspectives are offered to the readers. Expert opinion: Renal IRI is caused by a sudden and temporary impairment of blood flow to the organ. Defining the underlying cellular cascades involved in IRI will assist us in the identification of novel interventional targets to attenuate IRI with the potential to improve transplantation outcomes. Targeting mitochondrial function and cellular bioenergetics upstream of cellular damage may offer several advantages compared to targeting downstream inflammatory and fibrosis processes. An improved understanding of the cellular pathophysiological mechanisms leading to kidney injury will hopefully offer improved targeted therapies to prevent and treat the injury in the future.
Collapse
Affiliation(s)
- Nicolas Melis
- a Laboratory of Cellular and Molecular Biology , Center for Cancer Research, National Cancer Institute , Bethesda , MD , USA
| | - Raphael Thuillier
- b IRTOMIT , Inserm U1082 , Poitiers , France.,c Faculté de Médecine et de Pharmacie , Université de Poitiers , Poitiers , France.,d CHU Poitiers , Service de Biochimie , Poitiers , France.,e Fédération Hospitalo-Universitaire SUPORT , Poitiers , France
| | - Clara Steichen
- b IRTOMIT , Inserm U1082 , Poitiers , France.,c Faculté de Médecine et de Pharmacie , Université de Poitiers , Poitiers , France
| | - Sebastien Giraud
- b IRTOMIT , Inserm U1082 , Poitiers , France.,c Faculté de Médecine et de Pharmacie , Université de Poitiers , Poitiers , France.,d CHU Poitiers , Service de Biochimie , Poitiers , France
| | - Yse Sauvageon
- b IRTOMIT , Inserm U1082 , Poitiers , France.,c Faculté de Médecine et de Pharmacie , Université de Poitiers , Poitiers , France
| | - Jacques Kaminski
- b IRTOMIT , Inserm U1082 , Poitiers , France.,c Faculté de Médecine et de Pharmacie , Université de Poitiers , Poitiers , France
| | - Thomas Pelé
- b IRTOMIT , Inserm U1082 , Poitiers , France.,c Faculté de Médecine et de Pharmacie , Université de Poitiers , Poitiers , France
| | - Lionel Badet
- f Faculté de Médecine , Université Claude Bernard Lyon 1 , Villeurbanne , France.,g Hospices Civiles de Lyon , Service d'urologie et de chirurgie de la transplantation , Lyon , France
| | - Jean Pierre Richer
- b IRTOMIT , Inserm U1082 , Poitiers , France.,c Faculté de Médecine et de Pharmacie , Université de Poitiers , Poitiers , France.,h CHU de Poitiers , Service de chirurgie générale et endocrinienne , Poitiers , France.,i Faculté de Médecine et de Pharmacie , ABS Lab (Laboratoire d'Anatomie, Biomécanique et Simulation), Université de Poitiers , Poitiers , France
| | - Jonatan Barrera-Chimal
- j Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Medicina Traslacional , Instituto de Investigaciones Biomédicas, UNAM and Instituto Nacional de Cardiología Ignacio Chávez , Mexico City , Mexico
| | - Frédéric Jaisser
- k INSERM, UMRS 1138, Team 1 , Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris, Descartes University , Paris , France
| | - Michel Tauc
- l LP2M CNRS-UMR7370, LabEx ICST , Medical Faculty, Université Côte d'Azur , Nice , France
| | - Thierry Hauet
- b IRTOMIT , Inserm U1082 , Poitiers , France.,c Faculté de Médecine et de Pharmacie , Université de Poitiers , Poitiers , France.,d CHU Poitiers , Service de Biochimie , Poitiers , France.,e Fédération Hospitalo-Universitaire SUPORT , Poitiers , France.,i Faculté de Médecine et de Pharmacie , ABS Lab (Laboratoire d'Anatomie, Biomécanique et Simulation), Université de Poitiers , Poitiers , France.,m IBiSA Plateforme 'plate-forme MOdélisation Préclinique - Innovation Chirurgicale et Technologique (MOPICT)', Domaine Expérimental du Magneraud , Surgères , France
| |
Collapse
|
28
|
Muskiet MHA, Wheeler DC, Heerspink HJL. New pharmacological strategies for protecting kidney function in type 2 diabetes. Lancet Diabetes Endocrinol 2019; 7:397-412. [PMID: 30579729 DOI: 10.1016/s2213-8587(18)30263-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/16/2018] [Accepted: 08/21/2018] [Indexed: 12/31/2022]
Abstract
Type 2 diabetes is the leading cause of impaired kidney function, albuminuria, and renal replacement therapy globally, thus placing a large burden on health-care systems. Current treatment strategies rely on intensive glucose lowering as well as strict blood pressure control through blockade of the renin-angiotensin-aldosterone system. Such approaches might slow decline in kidney function, but many patients progress to end-stage kidney failure despite optimal therapy. In recent clinical trials, new-generation glucose-lowering drug classes, the sodium-glucose co-transporter-2 inhibitors and agents that target the incretin pathway, have been shown to improve kidney outcomes in patients with type 2 diabetes. Other new approaches, which have been developed on the basis of an improved understanding of the mechanisms that contribute to kidney damage in the context of diabetes, include use of drugs that block endothelin receptors (eg, atrasentan) and non-steroidal mineralocorticoid receptors (eg, finerenone). In this Review, we provide an overview of recent clinical data relevant to these new therapeutic approaches for management of kidney disease in the context of type 2 diabetes.
Collapse
Affiliation(s)
- Marcel H A Muskiet
- Diabetes Centre, Department of Internal Medicine, VU University Medical Centre, Amsterdam, Netherlands
| | - David C Wheeler
- Centre for Nephrology, University College London, London, UK.
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| |
Collapse
|
29
|
Adachi H, Kakuma T, Kawaguchi M, Kumagai E, Fukumoto Y. Effects of eplerenone on blood pressure and glucose metabolism in Japanese hypertensives with overweight or obesity. Medicine (Baltimore) 2019; 98:e14994. [PMID: 30985644 PMCID: PMC6485869 DOI: 10.1097/md.0000000000014994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE The impact of aldosterone blockade using eplerenone on hypertensives with obesity has not been clarified. We compared the efficacy and safety between eplerenone and trichlormethiazide in hypertensives with overweight or obesity. METHODS A prospective, randomized, open-labeled, blinded-endpoint design, multicenter trial enrolled 204 hypertension-treated outpatients with obesity [body mass index (BMI) ≥25 kg/m] evaluated by ambulatory blood pressure (BP) measurement. Patients were randomly assigned to receive 50 mg of eplerenone (n = 102) or 1 mg of trichlormethiazide (n = 102), each of which were administered once every morning. Primary efficacy endpoints were systolic and diastolic BPs and biomarkers of glucose metabolism after 6 months of treatment. RESULTS At baseline, BPs were comparable between the two groups. Systolic/diastolic blood pressure (SBP/DBP) were reduced from 153.9 ± 12.6/84.6 ± 11.8 to 129.8 ± 14.2/73.7 ± 12.2 mm Hg by eplerenone therapy and from 152.2 ± 12.5/85.2 ± 10.9 to 133.8 ± 12.6/76.1 ± 8.6 mm Hg by trichlormethiazide therapy (all; P < .001). The efficacy of SBP reduction after adjustment for age, sex, and BMI was significantly greater in the eplerenone group than the trichlormethiazide (P = .034), although the efficacy of DBP reduction was marginally significant (P = .072). Especially, the efficacy of BP reduction was more effective for aged over 65 years than less than 65 years. However, biomarkers of glucose metabolism were not significantly different between these 2 groups. CONCLUSION The eplerenone therapy was more effective in BP lowering in hypertensives with overweight or obesity than the trichlormethiazide therapy, especially in the elderly.
Collapse
Affiliation(s)
- Hisashi Adachi
- Department of Community Medicine, Kurume University School of Medicine
- Division of Cardio-Vascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | | | | | - Eita Kumagai
- Division of Cardio-Vascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Yoshihiro Fukumoto
- Division of Cardio-Vascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | | |
Collapse
|
30
|
|
31
|
Medzikovic L, de Vries CJM, de Waard V. NR4A nuclear receptors in cardiac remodeling and neurohormonal regulation. Trends Cardiovasc Med 2018; 29:429-437. [PMID: 30553703 DOI: 10.1016/j.tcm.2018.11.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/07/2018] [Accepted: 11/26/2018] [Indexed: 01/08/2023]
Abstract
Heart failure is characterized by the constant interplay between the underlying cardiac insult, degree of myocardial dysfunction and the activity of compensatory neurohormonal mechanisms. The sympathetic nervous system (SNS) and renin-angiotensin-aldosterone system (RAAS) become activated to maintain cardiac output; however, their chronic hyperactivity will eventually become deleterious. Several nuclear hormone receptors, including the mineralocorticoid receptor and estrogen receptor, are well-known to modulate cardiac disease. Recently, the subfamily of NR4A nuclear receptors i.e. Nur77, Nurr1 and NOR-1, are emerging as key players in cardiac stress responses, as well as pivotal regulators of neurohormonal mechanisms. In this review, we summarize current literature on NR4A nuclear receptors in the heart and in various components of the SNS, RAAS and immune system and discuss the functional implications for NR4As in cardiac function and disease.
Collapse
Affiliation(s)
- Lejla Medzikovic
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, Academic Medical Center, Amsterdam Cardiovascular Sciences, Meibergdreef 9, room K1-115, 1105 AZ Amsterdam, The Netherlands
| | - Carlie J M de Vries
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, Academic Medical Center, Amsterdam Cardiovascular Sciences, Meibergdreef 9, room K1-115, 1105 AZ Amsterdam, The Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, Academic Medical Center, Amsterdam Cardiovascular Sciences, Meibergdreef 9, room K1-115, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
32
|
Parker BM, Wertz SL, Pollard CM, Desimine VL, Maning J, McCrink KA, Lymperopoulos A. Novel Insights into the Crosstalk between Mineralocorticoid Receptor and G Protein-Coupled Receptors in Heart Adverse Remodeling and Disease. Int J Mol Sci 2018; 19:3764. [PMID: 30486399 PMCID: PMC6320977 DOI: 10.3390/ijms19123764] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/21/2018] [Accepted: 11/23/2018] [Indexed: 12/16/2022] Open
Abstract
The mineralocorticoid hormone aldosterone regulates sodium and potassium homeostasis but also adversely modulates the maladaptive process of cardiac adverse remodeling post-myocardial infarction. Through activation of its mineralocorticoid receptor (MR), a classic steroid hormone receptor/transcription factor, aldosterone promotes inflammation and fibrosis of the heart, the vasculature, and the kidneys. This is why MR antagonists reduce morbidity and mortality of heart disease patients and are part of the mainstay pharmacotherapy of advanced human heart failure. A plethora of animal studies using cell type⁻specific targeting of the MR gene have established the importance of MR signaling and function in cardiac myocytes, vascular endothelial and smooth muscle cells, renal cells, and macrophages. In terms of its signaling properties, the MR is distinct from nuclear receptors in that it has, in reality, two physiological hormonal agonists: not only aldosterone but also cortisol. In fact, in several tissues, including in the myocardium, cortisol is the primary hormone activating the MR. There is a considerable amount of evidence indicating that the effects of the MR in each tissue expressing it depend on tissue- and ligand-specific engagement of molecular co-regulators that either activate or suppress its transcriptional activity. Identification of these co-regulators for every ligand that interacts with the MR in the heart (and in other tissues) is of utmost importance therapeutically, since it can not only help elucidate fully the pathophysiological ramifications of the cardiac MR's actions, but also help design and develop novel better MR antagonist drugs for heart disease therapy. Among the various proteins the MR interacts with are molecules involved in cardiac G protein-coupled receptor (GPCR) signaling. This results in a significant amount of crosstalk between GPCRs and the MR, which can affect the latter's activity dramatically in the heart and in other cardiovascular tissues. This review summarizes the current experimental evidence for this GPCR-MR crosstalk in the heart and discusses its pathophysiological implications for cardiac adverse remodeling as well as for heart disease therapy. Novel findings revealing non-conventional roles of GPCR signaling molecules, specifically of GPCR-kinase (GRK)-5, in cardiac MR regulation are also highlighted.
Collapse
Affiliation(s)
- Barbara M Parker
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| | - Shelby L Wertz
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| | - Celina M Pollard
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| | - Victoria L Desimine
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| | - Jennifer Maning
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
- Present address: Jackson Memorial Hospital, Miami, FL 33136, USA.
| | - Katie A McCrink
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
- Present address: Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| |
Collapse
|
33
|
Lachaux M, Barrera-Chimal J, Nicol L, Rémy-Jouet I, Renet S, Dumesnil A, Wecker D, Richard V, Kolkhof P, Jaisser F, Ouvrard-Pascaud A, Mulder P. Short- and long-term administration of the non-steroidal mineralocorticoid receptor antagonist finerenone opposes metabolic syndrome-related cardio-renal dysfunction. Diabetes Obes Metab 2018; 20:2399-2407. [PMID: 29862614 DOI: 10.1111/dom.13393] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/23/2018] [Accepted: 05/25/2018] [Indexed: 12/22/2022]
Abstract
AIM To determine whether non-steroidal mineralocorticoid receptor (MR) antagonists oppose metabolic syndrome-related end-organ, i.e. cardiac, damage. MATERIALS AND METHODS In Zucker fa/fa rats, a rat model of metabolic syndrome, we assessed the effects of the non-steroidal MR antagonist finerenone (oral 2 mg/kg/day) on left ventricular (LV) function, haemodynamics and remodelling (using echocardiography, magnetic resonance imaging and biochemical methods). RESULTS Long-term (90 days) finerenone modified neither systolic blood pressure nor heart rate, but reduced LV end-diastolic pressure and LV end-diastolic pressure-volume relationship, without modifying LV end-systolic pressure and LV end-systolic pressure-volume relationship. Simultaneously, long-term finerenone reduced both LV systolic and diastolic diameters, associated with reductions in LV weight and LV collagen density, while proteinuria and renal nGAL expression were reduced. Short-term (7 days) finerenone improved LV haemodynamics and reduced LV systolic diameter, without modifying LV diastolic diameter. Moreover, short-term finerenone increased myocardial tissue perfusion and reduced myocardial reactive oxygen species, while plasma nitrite levels, an indicator of nitric oxide (NO) bio-availability, were increased. CONCLUSIONS In rats with metabolic syndrome, the non-steroidal MR antagonist finerenone opposed metabolic syndrome-related diastolic cardiac dysfunction and nephropathy. This involved acute effects, such as improved myocardial perfusion, reduced oxidative stress/increased NO bioavailability, as well as long-term effects, such as modifications in the myocardial structure.
Collapse
Affiliation(s)
- Marianne Lachaux
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | | | - Lionel Nicol
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | - Isabelle Rémy-Jouet
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | - Sylvanie Renet
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | - Anais Dumesnil
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | | | - Vincent Richard
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | | | - Frederic Jaisser
- Institut National de la Santé et de la Recherche Médicale U1138, Paris, France
| | - Antoine Ouvrard-Pascaud
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | - Paul Mulder
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| |
Collapse
|
34
|
Rodríguez-Lara SQ, García-Benavides L, Miranda-Díaz AG. The Renin-Angiotensin-Aldosterone System as a Therapeutic Target in Late Injury Caused by Ischemia-Reperfusion. Int J Endocrinol 2018; 2018:3614303. [PMID: 29849615 PMCID: PMC5904808 DOI: 10.1155/2018/3614303] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 01/09/2018] [Accepted: 02/07/2018] [Indexed: 12/19/2022] Open
Abstract
Ischemia-reperfusion (I/R) injury is a well-known phenomenon that involves different pathophysiological processes. Connection in diverse systems of survival brings about cellular dysfunction or even apoptosis. One of the survival systems of the cells, to the assault caused by ischemia, is the activation of the renin-angiotensin-aldosterone system (also known as an axis), which is focused on activating diverse signaling pathways to favor adaptation to the decrease in metabolic supports caused by the hypoxia. In trying to adapt to the I/R event, great changes occur that unchain cellular dysfunction with the capacity to lead to cell death, which translates into a poor prognosis due to the progression of dysfunction of the cellular activity. The search for the understanding of the diverse therapeutic alternatives in molecular coupling could favor the prognosis and evolution of patients who are subject to the I/R process.
Collapse
Affiliation(s)
- Simón Quetzalcóatl Rodríguez-Lara
- University of Guadalajara, Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Health Sciences Centre, Guadalajara, JAL, Mexico
| | - Leonel García-Benavides
- University of Guadalajara, Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Health Sciences Centre, Guadalajara, JAL, Mexico
| | - Alejandra Guillermina Miranda-Díaz
- University of Guadalajara, Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Health Sciences Centre, Guadalajara, JAL, Mexico
| |
Collapse
|
35
|
Infante M, Armani A, Mammi C, Fabbri A, Caprio M. Impact of Adrenal Steroids on Regulation of Adipose Tissue. Compr Physiol 2017; 7:1425-1447. [PMID: 28915330 DOI: 10.1002/cphy.c160037] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Corticosteroids are secreted by the adrenal glands and control the functions of adipose tissue via the activation of mineralocorticoid receptor (MR) and glucocorticoid receptor (GR). In turn, adipocytes release a large variety of adipokines into the bloodstream, regulating the function of several organs and tissues, including the adrenal glands, hereby controlling corticosteroid production. In adipose tissue, the activation of the MR by glucocorticoids (GC) and aldosterone affects important processes such as adipocyte differentiation, oxidative stress, autophagic flux, adipokine expression as well as local production of GC through upregulation of the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). Notably, the proinflammatory responses induced by the MR are counteracted by activation of the GR, whose activity inhibits the expression of inflammatory adipokines. Both GR and MR are deeply involved in adipogenesis and adipose expansion; hence pharmacological blockade of these two receptors has proven effective against adipose tissue dysfunction in experimental models of obesity and metabolic syndrome (MetS), suggesting a potential use for MR and GR antagonists in these clinical settings. Importantly, obesity and Cushing's syndrome (CS) share metabolic similarities and are characterized by high levels of circulating corticosteroids, which in turn are able to deeply affect adipose tissue. In addition, pharmacological approaches aimed at reducing aldosterone and GC levels, by means of the inhibition of CYP11B2 (aldosterone synthase) or 11β-HSD1, represent alternative strategies to counter the detrimental effects of excessive levels of corticosteroids, which are often observed in obesity and, more general, in MetS. © 2017 American Physiological Society. Compr Physiol 7:1425-1447, 2017.
Collapse
Affiliation(s)
- Marco Infante
- Unit of Endocrinology and Metabolic Diseases, Department of Systems Medicine, CTO A. Alesini Hospital, ASL Roma 2, University Tor Vergata, Rome, Italy
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Caterina Mammi
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Andrea Fabbri
- Unit of Endocrinology and Metabolic Diseases, Department of Systems Medicine, CTO A. Alesini Hospital, ASL Roma 2, University Tor Vergata, Rome, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| |
Collapse
|
36
|
Lee CY, Kuo WW, Baskaran R, Day CH, Pai PY, Lai CH, Chen YF, Chen RJ, Padma VV, Huang CY. Increased β-catenin accumulation and nuclear translocation are associated with concentric hypertrophy in cardiomyocytes. Cardiovasc Pathol 2017; 31:9-16. [PMID: 28802159 DOI: 10.1016/j.carpath.2017.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 01/19/2023] Open
Abstract
Defective Wnt/β-Catenin signaling, activated under various pathological conditions, can result in cardiac and vascular abnormalities. In the present study, the possible role of β-catenin over expression during cardiac hypertrophy was investigated. Ten samples from hearts of human patients with acute infarction, and granulation tissue from 20 patients and 10 from normal ones were collected in order to investigate roles of β-catenin in cardiac hypertrophy. H9c2 cardiomyoblast cells and Wistar rat primary neonatal cardiomyocytes were overexpressed with β-catenin. Expression levels of β-catenin protein were increased in human acute infarction tissues and rat hypertension heart tissues. Overexpression of this transcription factor induced actin filament formation and increased hypertrophic marker protein levels via MAPK pathway. In addition, β-catenin overexpression also resulted in increased elevation of NFATc3 and p-GATA4. Therefore, acute infarction resulted in β-catenin overexpression mediated hypertrophy in cardiomyocytes regulated through MAPK pathway.
Collapse
Affiliation(s)
- Cheng-Yu Lee
- Department of Cardiology, Taipei City Hospital, Zhongxiao Branch, Taipei, Taiwan
| | - Wei Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan
| | - Rathinasamy Baskaran
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | | | - Pei Ying Pai
- Division of Cardiology, China Medical University Hospital, Taichung 40447, Taiwan
| | - Chao Hung Lai
- Division of Cardiology, Department of Internal Medicine, Armed Force Taichung General Hospital, Taichung 41152, Taiwan
| | - Yu-Feng Chen
- Division of Cardiology, Department of Internal Medicine, Armed Force Taichung General Hospital, Taichung 41152, Taiwan
| | - Ray-Jade Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | | | - Chih Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan; Graduate Institute of Chinese Medical Science, China Medical University, Taichung 40402, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan; Faculty of Applied Sciences, Ton Duc Thang University, Tan Phong Ward, District 7, 700000 Ho Chi Minh City, Vietnam.
| |
Collapse
|
37
|
Ödek Ç, Kendirli T, Kocaay P, Azapağası E, Uçar T, Şıklar Z, Berberoğlu M. Acute reversible cardiomyopathy and heart failure in a child with acute adrenal crisis. Paediatr Int Child Health 2017; 37:148-151. [PMID: 27077627 DOI: 10.1080/20469047.2015.1120410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Acute adrenal crisis is a life-threatening disorder. Cardiovascular complications of the condition are usually limited to hypovolaemic hypotension and shock. An acute reversible cardiomyopathy and heart failure in association with acute adrenal crisis is rarely reported, particularly in children. A 6-year-old girl with adrenal crisis which was complicated by acute reversible cardiomyopathy is reported. Inotropic and ventilatory support in addition to intravenous hydrocortisone and furosemide therapy were required to achieve cardiovascular stability. The cardiomyopathy resolved over 5 days and she was discharged with normal cardiac and intellectual functions. Cardiomyopathy should be considered in patients with acute adrenal crisis demonstrating any symptoms or signs of heart failure.
Collapse
Affiliation(s)
- Çağlar Ödek
- a Divisions of Pediatric Critical Care, Department of Pediatrics, Faculty of Medicine, Ankara University , Ankara , Turkey
| | - Tanıl Kendirli
- a Divisions of Pediatric Critical Care, Department of Pediatrics, Faculty of Medicine, Ankara University , Ankara , Turkey
| | - Pınar Kocaay
- b Divisions of Pediatric Endocrinology, Department of Pediatrics, Faculty of Medicine , Ankara University , Ankara , Turkey
| | - Ebru Azapağası
- a Divisions of Pediatric Critical Care, Department of Pediatrics, Faculty of Medicine, Ankara University , Ankara , Turkey
| | - Tayfun Uçar
- c Divisions of Pediatric Cardiology, Department of Pediatrics, Faculty of Medicine , Ankara University , Ankara , Turkey
| | - Zeynep Şıklar
- b Divisions of Pediatric Endocrinology, Department of Pediatrics, Faculty of Medicine , Ankara University , Ankara , Turkey
| | - Merih Berberoğlu
- b Divisions of Pediatric Endocrinology, Department of Pediatrics, Faculty of Medicine , Ankara University , Ankara , Turkey
| |
Collapse
|
38
|
Sergeant CA, Africander D, Swart P, Swart AC. Sutherlandia frutescens modulates adrenal hormone biosynthesis, acts as a selective glucocorticoid receptor agonist (SEGRA) and displays anti-mineralocorticoid properties. JOURNAL OF ETHNOPHARMACOLOGY 2017; 202:290-301. [PMID: 28323049 DOI: 10.1016/j.jep.2017.03.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/10/2017] [Accepted: 03/14/2017] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sutherlandia frutescens is a traditional African medicinal plant used in the treatment of stress and anxiety, while also exhibiting anti-inflammatory properties. AIM OF STUDY The study aimed at linking anti-stress and anti-inflammatory properties of S. frutescens to its influence on glucocorticoid biosynthesis and the inflammatory response via steroid receptor interaction. MATERIALS AND METHODS The influence of S. frutescens extracts and sutherlandioside B (SUB),10 and 30µM, on key steroidogenic enzymes was assayed in COS-1 cells. Effects were also assayed on basal and stimulated hormone levels in the adrenal H295R cell model. Agonist activity for transactivation and transrepression of the extract and SUB with the glucocorticoid- (GR) and mineralocorticoid receptor (MR) was subsequently investigated. RESULTS Inhibitory effects of the extract towards progesterone conversion by CYP17A1 and CYP21A2 were significant. SUB inhibited CYP17A1 and 3β-HSD2, while not affecting CYP21A2. In H295R cells, SUB decreased cortisol and androgen precursors significantly. The extract decreased total steroid production (basal and stimulated) with cortisol and its precursor, deoxycortisol, together with mineralocorticoid metabolites significantly decreased under forskolin stimulated conditions. S. frutescens extracts and SUB repressed NF-κB-driven gene expression without activating GRE-driven gene expression and while neither activated MR mediated gene transcription, both antagonized the effects of aldosterone via the MR. CONCLUSION Data provide evidence linking anti-stress, anti-inflammatory and anti-hypertensive properties of S. frutescens to inhibition of steroidogenic enzymes and modulation of adrenal hormone biosynthesis. Findings suggesting S. frutescens and SUB exhibit dissociated glucocorticoid characteristics underline potential therapeutic applications in the treatment of inflammation and hypertension.
Collapse
Affiliation(s)
- C A Sergeant
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - D Africander
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - P Swart
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - A C Swart
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
39
|
Ruilope LM, Tamargo J. Renin–angiotensin system blockade: Finerenone. Nephrol Ther 2017; 13 Suppl 1:S47-S53. [DOI: 10.1016/j.nephro.2017.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 02/09/2017] [Indexed: 12/12/2022]
|
40
|
Richardson RV, Batchen EJ, Thomson AJW, Darroch R, Pan X, Rog-Zielinska EA, Wyrzykowska W, Scullion K, Al-Dujaili EAS, Diaz ME, Moran CM, Kenyon CJ, Gray GA, Chapman KE. Glucocorticoid receptor alters isovolumetric contraction and restrains cardiac fibrosis. J Endocrinol 2017; 232:437-450. [PMID: 28057868 PMCID: PMC5292999 DOI: 10.1530/joe-16-0458] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/05/2017] [Indexed: 01/17/2023]
Abstract
Corticosteroids directly affect the heart and vasculature and are implicated in the pathogenesis of heart failure. Attention is focussed upon the role of the mineralocorticoid receptor (MR) in mediating pro-fibrotic and other adverse effects of corticosteroids upon the heart. In contrast, the role of the glucocorticoid receptor (GR) in the heart and vasculature is less well understood. We addressed this in mice with cardiomyocyte and vascular smooth muscle deletion of GR (SMGRKO mice). Survival of SMGRKO mice to weaning was reduced compared with that of littermate controls. Doppler measurements of blood flow across the mitral valve showed an elongated isovolumetric contraction time in surviving adult SMGRKO mice, indicating impairment of the initial left ventricular contractile phase. Although heart weight was elevated in both genders, only male SMGRKO mice showed evidence of pathological cardiomyocyte hypertrophy, associated with increased myosin heavy chain-β expression. Left ventricular fibrosis, evident in both genders, was associated with elevated levels of mRNA encoding MR as well as proteins involved in cardiac remodelling and fibrosis. However, MR antagonism with spironolactone from birth only modestly attenuated the increase in pro-fibrotic gene expression in SMGRKO mice, suggesting that elevated MR signalling is not the primary driver of cardiac fibrosis in SMGRKO mice, and cardiac fibrosis can be dissociated from MR activation. Thus, GR contributes to systolic function and restrains normal cardiac growth, the latter through gender-specific mechanisms. Our findings suggest the GR:MR balance is critical in corticosteroid signalling in specific cardiac cell types.
Collapse
MESH Headings
- Animals
- Corticosterone/blood
- Female
- Fibrosis/metabolism
- Fibrosis/pathology
- Male
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocardial Contraction/genetics
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Nonmuscle Myosin Type IIB/genetics
- Nonmuscle Myosin Type IIB/metabolism
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Sex Factors
- Spironolactone/pharmacology
- Ventricular Function, Left/genetics
Collapse
Affiliation(s)
- Rachel V Richardson
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Emma J Batchen
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | | | - Rowan Darroch
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Xinlu Pan
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Eva A Rog-Zielinska
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Wiktoria Wyrzykowska
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Kathleen Scullion
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Emad A S Al-Dujaili
- DieteticsNutrition, and Biological Sciences Department, Queen Margaret University, Musselburgh, UK
| | - Mary E Diaz
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Carmel M Moran
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
- Edinburgh Preclinical ImagingUniversity of Edinburgh, Edinburgh, UK
| | - Christopher J Kenyon
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Gillian A Gray
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Karen E Chapman
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| |
Collapse
|
41
|
Avenatti E, Rebellato A, Iannaccone A, Battocchio M, Dassie F, Veglio F, Milan A, Fallo F. Left ventricular geometry and 24-h blood pressure profile in Cushing's syndrome. Endocrine 2017; 55:547-554. [PMID: 27179657 DOI: 10.1007/s12020-016-0986-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 05/06/2016] [Indexed: 12/17/2022]
Abstract
Cushing's syndrome (CS) is associated with cardiovascular disease. The impact of the hemodynamic load on left ventricular mass (LVM) in patients with CS is not well known. In fact, the effects of 24-h blood pressure (BP) load and BP circadian rhythm on cardiac structure and function have not been studied. Aim of the present study has thus been to assess the presence of cardiac remodeling in patients with newly diagnosed CS, combining evaluation of cardiac remodeling and assessment of BP burden derived by 24-h ambulatory blood pressure monitoring (ABPM). 25 patients (4 M, 21 F) with CS underwent echocardiography in order to assess cardiac morphology and geometry (relative wall thickness-RWT). As controls, 25 subjects similar for demographic characteristics and 24-h BP were used. CS Patients were similar to controls by age, sex, mean 24-h BP, and body mass index. There was a significant increase in left ventricular mass (LVM; 44.4 ± 14.7 vs. 36.9 ± 10 g/m2.7, p = 0.03) and a significant increase in RWT (0.46 ± 0.07 vs. 0.41 ± 0.08, p = 0.02) in CS patients compared to controls. The prevalence of CS patients with pressure non-dipping profile was greater than that of controls (56 vs. 16 %, p < 0.05), with no significant association with LVM or geometry. 24-h urinary cortisol was not associated with LVM (r = 0.1, p = 0.5) or RWT (r = 0.02, p = 0.89) in the CS group. In conclusion, LVM and the concentric pattern of the left ventricle are relatively independent from 24-h BP load and profile (dipping/non-dipping) in CS patients.
Collapse
Affiliation(s)
- Eleonora Avenatti
- Internal Medicine and Hypertension Division, Department of Medical Sciences, University Hospital AOU Città della Salute e della Scienza di Torino, University of Torino, Via Genova 3, 10126, Turin, Italy
| | - Andrea Rebellato
- Department of Medicine-DIMED, University of Padova, Padua, Italy
| | - Andrea Iannaccone
- Internal Medicine and Hypertension Division, Department of Medical Sciences, University Hospital AOU Città della Salute e della Scienza di Torino, University of Torino, Via Genova 3, 10126, Turin, Italy
| | | | - Francesca Dassie
- Department of Medicine-DIMED, University of Padova, Padua, Italy
| | - Franco Veglio
- Internal Medicine and Hypertension Division, Department of Medical Sciences, University Hospital AOU Città della Salute e della Scienza di Torino, University of Torino, Via Genova 3, 10126, Turin, Italy
| | - Alberto Milan
- Internal Medicine and Hypertension Division, Department of Medical Sciences, University Hospital AOU Città della Salute e della Scienza di Torino, University of Torino, Via Genova 3, 10126, Turin, Italy.
| | - Francesco Fallo
- Department of Medicine-DIMED, University of Padova, Padua, Italy
| |
Collapse
|
42
|
Ichige MHA, Pereira MG, Brum PC, Michelini LC. Experimental Evidences Supporting the Benefits of Exercise Training in Heart Failure. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 999:181-206. [PMID: 29022264 DOI: 10.1007/978-981-10-4307-9_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Heart Failure (HF), a common end point for many cardiovascular diseases, is a syndrome with a very poor prognosis. Although clinical trials in HF have achieved important outcomes in reducing mortality, little is known about functional mechanisms conditioning health improvement in HF patients. In parallel with clinical studies, basic science has been providing important discoveries to understand the mechanisms underlying the pathophysiology of HF, as well as to identify potential targets for the treatment of this syndrome. In spite of being the end-point of cardiovascular derangements caused by different etiologies, autonomic dysfunction, sympathetic hyperactivity, oxidative stress, inflammation and hormonal activation are common factors involved in the progression of this syndrome. Together these causal factors create a closed link between three important organs: brain, heart and the skeletal muscle. In the past few years, we and other groups have studied the beneficial effects of aerobic exercise training as a safe therapy to avoid the progression of HF. As summarized in this chapter, exercise training, a non-pharmacological tool without side effects, corrects most of the HF-induced neurohormonal and local dysfunctions within the brain, heart and skeletal muscles. These adaptive responses reverse oxidative stress, reduce inflammation, ameliorate neurohormonal control and improve both cardiovascular and skeletal muscle function, thus increasing the quality of life and reducing patients' morbimortality.
Collapse
Affiliation(s)
- Marcelo H A Ichige
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Marcelo G Pereira
- Department of Biodynamics of Human Body Movement, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Patrícia C Brum
- Department of Biodynamics of Human Body Movement, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil. .,National Institute for Science & Technology - INCT (In)activity & Exercise, CNPq - Niterói (RJ), Rio de Janeiro, Brazil.
| | - Lisete C Michelini
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.,National Institute for Science & Technology - INCT (In)activity & Exercise, CNPq - Niterói (RJ), Rio de Janeiro, Brazil
| |
Collapse
|
43
|
Chadwick JA, Bhattacharya S, Lowe J, Weisleder N, Rafael-Fortney JA. Renin-angiotensin-aldosterone system inhibitors improve membrane stability and change gene-expression profiles in dystrophic skeletal muscles. Am J Physiol Cell Physiol 2016; 312:C155-C168. [PMID: 27881412 PMCID: PMC5336592 DOI: 10.1152/ajpcell.00269.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 01/16/2023]
Abstract
Angiotensin-converting enzyme inhibitors (ACEi) and mineralocorticoid receptor (MR) antagonists are FDA-approved drugs that inhibit the renin-angiotensin-aldosterone system (RAAS) and are used to treat heart failure. Combined treatment with the ACEi lisinopril and the nonspecific MR antagonist spironolactone surprisingly improves skeletal muscle, in addition to heart function and pathology in a Duchenne muscular dystrophy (DMD) mouse model. We recently demonstrated that MR is present in all limb and respiratory muscles and functions as a steroid hormone receptor in differentiated normal human skeletal muscle fibers. The goals of the current study were to begin to define cellular and molecular mechanisms mediating the skeletal muscle efficacy of RAAS inhibitor treatment. We also compared molecular changes resulting from RAAS inhibition with those resulting from the current DMD standard-of-care glucocorticoid treatment. Direct assessment of muscle membrane integrity demonstrated improvement in dystrophic mice treated with lisinopril and spironolactone compared with untreated mice. Short-term treatments of dystrophic mice with specific and nonspecific MR antagonists combined with lisinopril led to overlapping gene-expression profiles with beneficial regulation of metabolic processes and decreased inflammatory gene expression. Glucocorticoids increased apoptotic, proteolytic, and chemokine gene expression that was not changed by RAAS inhibitors in dystrophic mice. Microarray data identified potential genes that may underlie RAAS inhibitor treatment efficacy and the side effects of glucocorticoids. Direct effects of RAAS inhibitors on membrane integrity also contribute to improved pathology of dystrophic muscles. Together, these data will inform clinical development of MR antagonists for treating skeletal muscles in DMD.
Collapse
Affiliation(s)
- Jessica A Chadwick
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio; and
| | - Sayak Bhattacharya
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio; and.,Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Jeovanna Lowe
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio; and
| | - Noah Weisleder
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio; and.,Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Jill A Rafael-Fortney
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio; and
| |
Collapse
|
44
|
Heggermont WA, Goethals M, Dierckx R, Verstreken S, Bartunek J, Vanderheyden M. Should MRAs be at the front row in heart failure? A plea for the early use of mineralocorticoid receptor antagonists in medical therapy for heart failure based on clinical experience. Heart Fail Rev 2016; 21:699-701. [PMID: 27620301 DOI: 10.1007/s10741-016-9583-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The brand new 2016 ESC guidelines for the treatment of acute and chronic heart failure continue to give a prominent place to mineralocorticoid receptor antagonists in the treatment of chronic heart failure with reduced ejection fraction (HFrEF). In the prevention of HF hospitalization and death, a class I, level of recommendation A, is given to MRAs for patients with HFrEF, who remain symptomatic despite treatment with an ACE-inhibitor and a beta-blocker and have an LVEF below 35 %. This recommendation is primarily based on two landmark trials, the RALES trial (for spironolactone) and the EMPHASIS-HF trial (for eplerenone). A crucial question is, however, why MRAs are advised only in "third place," i.e., after optimal up-titration of ACE-inhibitors and beta-blockers. We wonder whether MRAs could not or should not be given earlier in the treatment of HFrEF, namely before or together with the up-titration of ACE-inhibitors and beta-blockers. Several arguments to support this plea are described in this short paper.
Collapse
Affiliation(s)
- Ward A Heggermont
- Cardiovascular Center, OLV Ziekenhuis Aalst, Moorselbaan 164, 9300, Aalst, Belgium.
| | - Marc Goethals
- Cardiovascular Center, OLV Ziekenhuis Aalst, Moorselbaan 164, 9300, Aalst, Belgium
| | - Riet Dierckx
- Cardiovascular Center, OLV Ziekenhuis Aalst, Moorselbaan 164, 9300, Aalst, Belgium
| | - Sofie Verstreken
- Cardiovascular Center, OLV Ziekenhuis Aalst, Moorselbaan 164, 9300, Aalst, Belgium
| | - Jozef Bartunek
- Cardiovascular Center, OLV Ziekenhuis Aalst, Moorselbaan 164, 9300, Aalst, Belgium
| | - Marc Vanderheyden
- Cardiovascular Center, OLV Ziekenhuis Aalst, Moorselbaan 164, 9300, Aalst, Belgium
| |
Collapse
|
45
|
Okoshi MP, Cezar MDM, Iyomasa RM, Silva MB, Costa LCO, Martinez PF, Campos DHS, Damatto RL, Minicucci MF, Cicogna AC, Okoshi K. Effects of early aldosterone antagonism on cardiac remodeling in rats with aortic stenosis-induced pressure overload. Int J Cardiol 2016; 222:569-575. [PMID: 27513653 DOI: 10.1016/j.ijcard.2016.07.266] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 07/29/2016] [Accepted: 07/30/2016] [Indexed: 11/17/2022]
Abstract
UNLABELLED Aldosterone plays a pivotal role in the pathophysiology of systolic heart failure. However, whether early aldosterone antagonism improves cardiac remodeling during persistent pressure overload is unsettled. We evaluated the effects of aldosterone antagonist spironolactone on cardiac remodeling in rats with ascending aortic stenosis (AS). METHODS Three days after inducing AS, weaning rats were randomized to receive spironolactone (AS-SPR, 20mg/kg/day) or no drug (AS) for 18weeks, and compared with sham-operated rats. Myocardial function was studied in isolated left ventricular (LV) papillary muscles. STATISTICAL ANALYSES ANOVA or Kruskal-Wallis tests. RESULTS Echocardiogram showed that LV diastolic (Sham 8.73±0.57; AS 8.30±1.10; AS-SPR 9.19±1.15mm) and systolic (Sham 4.57±0.67; AS 3.61±1.49; AS-SPR 4.62±1.48mm) diameters, left atrial diameter (Sham 5.80±0.44; AS 7.15±1.22; AS-SPR 8.02±1.17mm), and LV mass were higher in AS-SPR than AS. Posterior wall shortening velocity (Sham 38.5±3.8; AS 35.6±5.6; AS-SPR 31.1±3.8mm/s) was lower in AS-SPR than Sham and AS; E/A ratio was higher in AS-SPR than Sham. Developed tension was lower in AS and AS-SPR than Sham. Time to peak tension was higher in AS-SPR than Sham and AS after post-rest contraction. Right ventricle weight was higher in AS-SPR than AS, suggesting more severe heart failure in AS-SPR than AS. Interstitial collagen fractional area and myocardial hydroxyproline concentration were higher in AS than Sham. Metalloproteinase-2 and -9 activity, evaluated by zymography, did not differ between groups. CONCLUSION Early spironolactone administration causes further hypertrophy in cardiac chambers, and left ventricular dilation and dysfunction in rats with AS-induced chronic pressure overload.
Collapse
Affiliation(s)
- M P Okoshi
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University, UNESP, Brazil.
| | - M D M Cezar
- Itapeva Social and Agrarian Sciences College, FAIT, Itapeva, SP, Brazil
| | - R M Iyomasa
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University, UNESP, Brazil
| | - M B Silva
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University, UNESP, Brazil
| | - L C O Costa
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University, UNESP, Brazil
| | - P F Martinez
- Federal University of Mato Grosso do Sul, Campo Grande, Brazil
| | - D H S Campos
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University, UNESP, Brazil
| | - R L Damatto
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University, UNESP, Brazil; Itapeva Social and Agrarian Sciences College, FAIT, Itapeva, SP, Brazil
| | - M F Minicucci
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University, UNESP, Brazil
| | - A C Cicogna
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University, UNESP, Brazil
| | - K Okoshi
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University, UNESP, Brazil
| |
Collapse
|
46
|
Nguyen Dinh Cat A, Antunes TT, Callera GE, Sanchez A, Tsiropoulou S, Dulak-Lis MG, Anagnostopoulou A, He Y, Montezano AC, Jaisser F, Touyz RM. Adipocyte-Specific Mineralocorticoid Receptor Overexpression in Mice Is Associated With Metabolic Syndrome and Vascular Dysfunction: Role of Redox-Sensitive PKG-1 and Rho Kinase. Diabetes 2016; 65:2392-403. [PMID: 27207514 DOI: 10.2337/db15-1627] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 04/09/2016] [Indexed: 11/13/2022]
Abstract
Mineralocorticoid receptor (MR) expression is increased in adipose tissue from obese individuals and animals. We previously demonstrated that adipocyte-MR overexpression (Adipo-MROE) in mice is associated with metabolic changes. Whether adipocyte MR directly influences vascular function in these mice is unknown. We tested this hypothesis in resistant mesenteric arteries from Adipo-MROE mice using myography and in cultured adipocytes. Molecular mechanisms were probed in vessels/vascular smooth muscle cells and adipose tissue/adipocytes and focused on redox-sensitive pathways, Rho kinase activity, and protein kinase G type-1 (PKG-1) signaling. Adipo-MROE versus control-MR mice exhibited reduced vascular contractility, associated with increased generation of adipocyte-derived hydrogen peroxide, activation of vascular redox-sensitive PKG-1, and downregulation of Rho kinase activity. Associated with these vascular changes was increased elastin content in Adipo-MROE. Inhibition of PKG-1 with Rp-8-Br-PET-cGMPS normalized vascular contractility in Adipo-MROE. In the presence of adipocyte-conditioned culture medium, anticontractile effects of the adipose tissue were lost in Adipo-MROE mice but not in control-MR mice. In conclusion, adipocyte-MR upregulation leads to impaired contractility with preserved endothelial function and normal blood pressure. Increased elasticity may contribute to hypocontractility. We also identify functional cross talk between adipocyte MR and arteries and describe novel mechanisms involving redox-sensitive PKG-1 and Rho kinase. Our results suggest that adipose tissue from Adipo-MROE secrete vasoactive factors that preferentially influence vascular smooth muscle cells rather than endothelial cells. Our findings may be important in obesity/adiposity where adipocyte-MR expression/signaling is amplified and vascular risk increased.
Collapse
Affiliation(s)
- Aurelie Nguyen Dinh Cat
- Cardiovascular Research and Medical Sciences Institute, University of Glasgow, Glasgow, U.K.
| | - Tayze T Antunes
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Glaucia E Callera
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Ana Sanchez
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Sofia Tsiropoulou
- Cardiovascular Research and Medical Sciences Institute, University of Glasgow, Glasgow, U.K
| | - Maria G Dulak-Lis
- Cardiovascular Research and Medical Sciences Institute, University of Glasgow, Glasgow, U.K
| | | | - Ying He
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Augusto C Montezano
- Cardiovascular Research and Medical Sciences Institute, University of Glasgow, Glasgow, U.K
| | - Frederic Jaisser
- INSERM Unit 1138 Team 1, Centre de Recherche des Cordeliers, University Pierre and Marie Curie, Paris, France INSERM, Clinical Investigation Centre 1430, APHP, Henri Mondor Hospital, Pole VERDI, Paris East University, Creteil, France
| | - Rhian M Touyz
- Cardiovascular Research and Medical Sciences Institute, University of Glasgow, Glasgow, U.K. Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
47
|
Ibrahim SIA, Strong JA, Zhang JM. Mineralocorticoid Receptor, A Promising Target for Improving Management of Low Back Pain by Epidural Steroid Injections. ACTA ACUST UNITED AC 2016; 3:177-184. [PMID: 28956026 PMCID: PMC5611848 DOI: 10.24015/japm.2016.0023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIM OF REVIEW Low back pain is a major health problem in United States and worldwide. In this review, we aim to show that mineralocorticoid receptor (MR) activation has a critical role in the initiation of immune and inflammatory responses, which in turn can impact the effectiveness of the currently used steroids for epidural injections in low back pain management since most steroids activate MR in addition to the primary target, glucocorticoid receptor (GR). Moreover, we would like to determine some of the benefits of blocking the MR-induced negative effects. Overall, we propose a novel therapeutic approach for low back pain management by using a combination of a MR antagonist and a GR agonist in the epidural injections. METHOD We will first introduce the societal cost of low back pain and discuss how epidural steroid injections became a popular treatment for this condition. We will then describe several preclinical models used for the study of low back pain conditions and the findings with respect to the role of MR in the development of inflammatory low back pain. RECENT FINDINGS MR has pro-inflammatory effects in many tissues which can counteract the anti-inflammatory effects induced by GR activation. Blocking MR using the selective MR antagonist eplerenone can reduce pain and sensory neuron excitability in experimental models of low back pain. Moreover, combining the MR antagonist with clinically used steroids is more effective in reducing pain behaviors than using the steroids alone. SUMMARY MR antagonists are promising candidates to increase the effectiveness of currently used steroids. Since the activation of the MR is evident in preclinical models of low back pain, blocking its deleterious effects can be beneficial in managing inflammatory pain conditions.
Collapse
Affiliation(s)
- Shaimaa I A Ibrahim
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, USA.,Graduate Program in Molecular, Cellular, and Biochemical Pharmacology, University of Cincinnati, Cincinnati, USA
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, USA
| |
Collapse
|
48
|
Cruz-Topete D, Myers PH, Foley JF, Willis MS, Cidlowski JA. Corticosteroids Are Essential for Maintaining Cardiovascular Function in Male Mice. Endocrinology 2016; 157:2759-71. [PMID: 27219275 PMCID: PMC4929548 DOI: 10.1210/en.2015-1604] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activation of the hypothalamic-pituitary-adrenal axis results in the release of hormones from the adrenal glands, including glucocorticoids and mineralocorticoids. The physiological association between corticosteroids and cardiac disease is becoming increasingly recognized; however, the mechanisms underlying this association are not well understood. To determine the biological effects of corticosteroids on the heart, we investigated the impact of adrenalectomy in C57BL/6 male mice. Animals were adrenalectomized (ADX) at 1 month of age and maintained for 3-6 months after surgery to evaluate the effects of long-term adrenalectomy on cardiac function. Morphological evaluation suggested that ADX mice showed significantly enlarged hearts compared with age-matched intact controls. These changes in morphology correlated with deficits in left ventricular (LV) function and electrocardiogram (ECG) abnormalities in ADX mice. Correlating with these functional defects, gene expression analysis of ADX hearts revealed aberrant expression of a large cohort of genes associated with cardiac hypertrophy and arrhythmia. Combined corticosterone and aldosterone replacement treatment prevented the emergence of cardiac abnormalities in ADX mice, whereas corticosterone replacement prevented the effects of adrenalectomy on LV function but did not block the emergence of ECG alterations. Aldosterone replacement did not preserve the LV function but prevented ECG abnormalities. Together, the data indicate that adrenal glucocorticoids and mineralocorticoids either directly or indirectly have selective effects in the heart and their signaling pathways are essential in maintaining normal cardiac function.
Collapse
Affiliation(s)
- Diana Cruz-Topete
- Signal Transduction Laboratory (D.C.-T., J.A.C.), Comparative Medicine Branch (P.H.M.), and Cellular and Molecular Pathology Branch (J.F.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland 20892; and McAllister Heart Institute (M.S.W.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514
| | - Page H Myers
- Signal Transduction Laboratory (D.C.-T., J.A.C.), Comparative Medicine Branch (P.H.M.), and Cellular and Molecular Pathology Branch (J.F.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland 20892; and McAllister Heart Institute (M.S.W.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514
| | - Julie F Foley
- Signal Transduction Laboratory (D.C.-T., J.A.C.), Comparative Medicine Branch (P.H.M.), and Cellular and Molecular Pathology Branch (J.F.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland 20892; and McAllister Heart Institute (M.S.W.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514
| | - Monte S Willis
- Signal Transduction Laboratory (D.C.-T., J.A.C.), Comparative Medicine Branch (P.H.M.), and Cellular and Molecular Pathology Branch (J.F.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland 20892; and McAllister Heart Institute (M.S.W.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514
| | - John A Cidlowski
- Signal Transduction Laboratory (D.C.-T., J.A.C.), Comparative Medicine Branch (P.H.M.), and Cellular and Molecular Pathology Branch (J.F.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland 20892; and McAllister Heart Institute (M.S.W.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514
| |
Collapse
|
49
|
Sabourin J, Bartoli F, Antigny F, Gomez AM, Benitah JP. Transient Receptor Potential Canonical (TRPC)/Orai1-dependent Store-operated Ca2+ Channels: NEW TARGETS OF ALDOSTERONE IN CARDIOMYOCYTES. J Biol Chem 2016; 291:13394-409. [PMID: 27129253 DOI: 10.1074/jbc.m115.693911] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Indexed: 12/31/2022] Open
Abstract
Store-operated Ca(2+) entry (SOCE) has emerged as an important mechanism in cardiac pathology. However, the signals that up-regulate SOCE in the heart remain unexplored. Clinical trials have emphasized the beneficial role of mineralocorticoid receptor (MR) signaling blockade in heart failure and associated arrhythmias. Accumulated evidence suggests that the mineralocorticoid hormone aldosterone, through activation of its receptor, MR, might be a key regulator of Ca(2+) influx in cardiomyocytes. We thus assessed whether and how SOCE involving transient receptor potential canonical (TRPC) and Orai1 channels are regulated by aldosterone/MR in neonatal rat ventricular cardiomyocytes. Molecular screening using qRT-PCR and Western blotting demonstrated that aldosterone treatment for 24 h specifically increased the mRNA and/or protein levels of Orai1, TRPC1, -C4, -C5, and stromal interaction molecule 1 through MR activation. These effects were correlated with a specific enhancement of SOCE activities sensitive to store-operated channel inhibitors (SKF-96365 and BTP2) and to a potent Orai1 blocker (S66) and were prevented by TRPC1, -C4, and Orai1 dominant negative mutants or TRPC5 siRNA. A mechanistic approach showed that up-regulation of serum- and glucocorticoid-regulated kinase 1 mRNA expression by aldosterone is involved in enhanced SOCE. Functionally, 24-h aldosterone-enhanced SOCE is associated with increased diastolic [Ca(2+)]i, which is blunted by store-operated channel inhibitors. Our study provides the first evidence that aldosterone promotes TRPC1-, -C4-, -C5-, and Orai1-mediated SOCE in cardiomyocytes through an MR and serum- and glucocorticoid-regulated kinase 1 pathway.
Collapse
Affiliation(s)
- Jessica Sabourin
- From the UMR S1180, INSERM, Université Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France and
| | - Fiona Bartoli
- From the UMR S1180, INSERM, Université Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France and
| | - Fabrice Antigny
- UMR S999, INSERM, Université Paris-Sud, Université Paris-Saclay, Centre Chirurgical Marie Lannelongue, 92350 Le Plessis Robinson, France
| | - Ana Maria Gomez
- From the UMR S1180, INSERM, Université Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France and
| | - Jean-Pierre Benitah
- From the UMR S1180, INSERM, Université Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France and
| |
Collapse
|
50
|
Adult nephron-specific MR-deficient mice develop a severe renal PHA-1 phenotype. Pflugers Arch 2016; 468:895-908. [PMID: 26762397 DOI: 10.1007/s00424-015-1785-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 12/21/2015] [Accepted: 12/27/2015] [Indexed: 11/27/2022]
Abstract
Aldosterone is the main mineralocorticoid hormone controlling sodium balance, fluid homeostasis, and blood pressure by regulating sodium reabsorption in the aldosterone-sensitive distal nephron (ASDN). Germline loss-of-function mutations of the mineralocorticoid receptor (MR) in humans and in mice lead to the "renal" form of type 1 pseudohypoaldosteronism (PHA-1), a case of aldosterone resistance characterized by salt wasting, dehydration, failure to thrive, hyperkalemia, and metabolic acidosis. To investigate the importance of MR in adult epithelial cells, we generated nephron-specific MR knockout mice (MR(Pax8/LC1)) using a doxycycline-inducible system. Under standard diet, MR(Pax8/LC1) mice exhibit inability to gain weight and significant weight loss compared to control mice. Interestingly, despite failure to thrive, MR(Pax8/LC1) mice survive but develop a severe PHA-1 phenotype with higher urinary Na(+) levels, decreased plasma Na(+), hyperkalemia, and higher levels of plasma aldosterone. This phenotype further worsens and becomes lethal under a sodium-deficient diet. Na(+)/Cl(-) co-transporter (NCC) protein expression and its phosphorylated form are downregulated in the MR(Pax8/LC1) knockouts, as well as the αENaC protein expression level, whereas the expression of glucocorticoid receptor (GR) is increased. A diet rich in Na(+) and low in K(+) does not restore plasma aldosterone to control levels but is sufficient to restore body weight, plasma, and urinary electrolytes. In conclusion, MR deletion along the nephron fully recapitulates the features of severe human PHA-1. ENaC protein expression is dependent on MR activity. Suppression of NCC under hyperkalemia predominates in a hypovolemic state.
Collapse
|