1
|
Kunz S, Meng Y, Schneider H, Brunnenkant L, Höhne M, Kühnle T, Reincke M, Theodoropoulou M, Bidlingmaier M. Fast and reliable quantification of aldosterone, cortisol and cortisone via LC-MS/MS to study 11β-hydroxysteroid dehydrogenase activities in primary cell cultures. J Steroid Biochem Mol Biol 2024; 244:106610. [PMID: 39214289 DOI: 10.1016/j.jsbmb.2024.106610] [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: 03/20/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Cell culture experiments can support characterization of enzymatic activities in healthy and tumorous human tissues. Liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) enables simultaneous measurement of several steroids from a single sample, facilitating analysis of molecular pathways involved in steroid biosynthesis. We developed a reliable but fast method for quantification of cortisol, cortisone and aldosterone in cell culture supernatant. Validation, including investigation of matrix-matched calibration, was performed for two different cell types. Utility of the method was demonstrated in the study of 11β-hydroxysteroid dehydrogenase type 2 (HSD11B2) activity under conditions of glucocorticoid and mineralocorticoid excess in different cell types. Aldosterone, cortisol and cortisone were extracted by liquid-liquid extraction (LLE) with methyl tert-butyl ether from 1 mL of cell culture supernatant. Steroids were separated on a Kinetex biphenyl column (50 ×2.1 mm, 2.6 µm) with gradient elution of water and methanol containing 2 mM ammonium format and analysed in multiple reaction monitoring mode after positive electrospray ionization. Application of the method included cell culture experiments with two different primary cell types, human coronary artery smooth muscle cells (HCSMC) and human coronary artery endothelial cells (EC). Cells were treated with different concentrations of cortisol, aldosterone and mifepristone, a glucocorticoid receptor antagonist and quantitative PCR was performed. The method exhibits high precision (CV ≤ 6 %) and accuracy (deviation from nominal concentration ≤ 6 %) for concentrations above the limit of quantification (LoQ) which is 0.11, 0.56 and 0.69 nmol/L for aldosterone, cortisone and cortisol, respectively. Calibration curves did not differ when prepared in media or solvent. The method enabled us to confirm activity of HSD11B2 and concentration dependent conversion of cortisol to cortisone in HCSMC (median conversion ratio at 140 nM cortisol = 1.46 %). In contrast we did not observe any HSD11B2 activity in EC. Neither addition of high aldosterone, nor addition of 1 µM mifepristone had impact on glucocorticoid concentrations. Quantitative PCR revealed expression of HSD11B1 and HSD11B2 in HCSMC but not in EC. We present a fast and reliable method for quantification of cortisol, cortisone and aldosterone in cell culture supernatants. The method enabled us to study HSD11B2 activity in two different cell types and will support future experiments investigating mechanisms of target organ damage in conditions of glucocorticoid and mineralocorticoid excess.
Collapse
Affiliation(s)
- Sonja Kunz
- Department of Medicine IV, LMU University Hospital, LMU Munich, Ziemssenstraße 5, Munich 80336, Germany.
| | - Yao Meng
- Department of Medicine IV, LMU University Hospital, LMU Munich, Ziemssenstraße 5, Munich 80336, Germany; Department of geriatric medicine, Gan Su provincial hospital, Dong Gang West Road 204, Lan Zhou 731100, China.
| | - Holger Schneider
- Department of Medicine IV, LMU University Hospital, LMU Munich, Ziemssenstraße 5, Munich 80336, Germany.
| | - Laura Brunnenkant
- Department of Medicine IV, LMU University Hospital, LMU Munich, Ziemssenstraße 5, Munich 80336, Germany.
| | - Michaela Höhne
- Department of Medicine IV, LMU University Hospital, LMU Munich, Ziemssenstraße 5, Munich 80336, Germany.
| | - Tim Kühnle
- Department of Medicine IV, LMU University Hospital, LMU Munich, Ziemssenstraße 5, Munich 80336, Germany.
| | - Martin Reincke
- Department of Medicine IV, LMU University Hospital, LMU Munich, Ziemssenstraße 5, Munich 80336, Germany.
| | - Marily Theodoropoulou
- Department of Medicine IV, LMU University Hospital, LMU Munich, Ziemssenstraße 5, Munich 80336, Germany.
| | - Martin Bidlingmaier
- Department of Medicine IV, LMU University Hospital, LMU Munich, Ziemssenstraße 5, Munich 80336, Germany.
| |
Collapse
|
2
|
Wang Z, Li L, Yang S, Li Z, Zhang P, Shi R, Zhou X, Tang X, Li Q. Possible mechanisms of SARS-CoV-2-associated myocardial fibrosis: reflections in the post-pandemic era. Front Microbiol 2024; 15:1470953. [PMID: 39444690 PMCID: PMC11497467 DOI: 10.3389/fmicb.2024.1470953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Since December 2019, coronavirus disease 2019 (COVID-19) has been spreading worldwide with devastating immediate or long-term effects on people's health. Although the lungs are the primary organ affected by COVID-19, individuals infected with SARS-CoV-2 also develop systemic lesions involving multiple organs throughout the body, such as the cardiovascular system. Emerging evidence reveals that COVID-19 could generate myocardial fibrosis, termed "COVID-19-associated myocardial fibrosis." It can result from the activation of fibroblasts via the renin-angiotensin-aldosterone system (RAAS), transforming growth factor-β1 (TGF-β1), microRNAs, and other pathways, and can also occur in other cellular interactions with SARS-CoV-2, such as immunocytes, endothelial cells. Nonetheless, to gain a more profound insight into the natural progression of COVID-19-related myocardial fibrosis, additional investigations are necessary. This review delves into the underlying mechanisms contributing to COVID-19-associated myocardial fibrosis while also examining the antifibrotic potential of current COVID-19 treatments, thereby offering guidance for future clinical trials of these medications. Ultimately, we propose future research directions for COVID-19-associated myocardial fibrosis in the post-COVID-19 era, such as artificial intelligence (AI) telemedicine. We also recommend that relevant tests be added to the follow-up of COVID-19 patients to detect myocardial fibrosis promptly.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Luwei Li
- Department of Pediatric Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Third Clinical Medical College of Zhengzhou University, Zhengzhou, China
| | - Shuai Yang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojuan Tang
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Li
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Escudero DS, Fantinelli JC, Martínez VR, González Arbeláez LF, Amarillo ME, Pérez NG, Díaz RG. Hydrocortisone cardioprotection in ischaemia/reperfusion injury involves antioxidant mechanisms. Eur J Clin Invest 2024; 54:e14172. [PMID: 38293760 DOI: 10.1111/eci.14172] [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: 10/27/2023] [Revised: 12/22/2023] [Accepted: 01/12/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND Glucocorticoid (GR) and mineralocorticoid (MR) receptors are highly expressed in cardiac tissue, and both can be activated by corticosteroids. MR activation, in acute myocardial infarction (AMI), worsens cardiac function, and increase NHE activity contributing to the deleterious process. In contrast, effects of GR activation are not fully understood, probably because of the controversial scenario generated by using different doses or potencies of corticosteroids. AIMS We tested the hypothesis that an acute dose of hydrocortisone (HC), a low-potency glucocorticoid, in a murine model of AMI could be cardioprotective by regulating NHE1 activity, leading to a decrease in oxidative stress. MATERIALS AND METHODS Isolated hearts from Wistar rats were subjected to regional ischemic protocol. HC (10 nmol/L) was added to the perfusate during early reperfusion. Infarct size and oxidative stress were determined. Isolated papillary muscles from non-infarcted hearts were used to evaluate HC effect on sodium-proton exchanger 1 (NHE1) by analysing intracellular pH recovery from acute transient acidosis. RESULTS HC treatment decreased infarct size, improved cardiac mechanics, reduced oxidative stress after AMI, while restoring the decreased level of the pro-fusion mitochondrial protein MFN-2. Co-treatment with the GR-blocker Mifepristone avoided these effects. HC reduced NHE1 activity by increasing the NHE1 pro-inhibiting Ser648 phosphorylation site and its upstream kinase AKT. HC restored the decreased AKT phosphorylation and anti-apoptotic BCL-2 protein expression detected after AMI. CONCLUSIONS Our results provide the first evidence that acute HC treatment during early reperfusion induces cardioprotection against AMI, associated with a non-genomic HC-triggered NHE1 inhibition by AKT and antioxidant action that might involves mitochondrial dynamics improvement.
Collapse
Affiliation(s)
- Daiana S Escudero
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Established Investigator of Comisión de Investigaciones Científicas (CIC), Buenos Aires, Argentina
| | - Juliana C Fantinelli
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Established Investigators of Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Valeria R Martínez
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Established Investigators of Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Luisa F González Arbeláez
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Established Investigators of Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María E Amarillo
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Fellow of Agencia Nacional de Promoción Científica y Tecnológica (Agencia I+D+i), Buenos Aires, Argentina
| | - Néstor G Pérez
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Established Investigators of Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Romina G Díaz
- Centro de Investigaciones Cardiovasculares 'Dr. Horacio E. Cingolani', Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, La Plata, Argentina
- Established Investigators of Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
4
|
Deng L, Pollmeier L, Bednarz R, Cao C, Laurette P, Wirth L, Mamazhakypov A, Bode C, Hein L, Gilsbach R, Lother A. Atlas of cardiac endothelial cell enhancer elements linking the mineralocorticoid receptor to pathological gene expression. SCIENCE ADVANCES 2024; 10:eadj5101. [PMID: 38446896 PMCID: PMC10917356 DOI: 10.1126/sciadv.adj5101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/31/2024] [Indexed: 03/08/2024]
Abstract
Endothelial cells play crucial roles in physiology and are increasingly recognized as therapeutic targets in cardiovascular disease. Here, we analyzed the regulatory landscape of cardiac endothelial cells by assessing chromatin accessibility, histone modifications, and 3D chromatin organization and confirmed the functional relevance of enhancer-promoter interactions by CRISPRi-mediated enhancer silencing. We used this dataset to explore mechanisms of transcriptional regulation in cardiovascular disease and compared six different experimental models of heart failure, hypertension, or diabetes. Enhancers that regulate gene expression in diseased endothelial cells were enriched with binding sites for a distinct set of transcription factors, including the mineralocorticoid receptor (MR), a known drug target in heart failure and hypertension. For proof of concept, we applied endothelial cell-specific MR deletion in mice to confirm MR-dependent gene expression and predicted direct MR target genes. Overall, we have compiled here a comprehensive atlas of cardiac endothelial cell enhancer elements that provides insight into the role of transcription factors in cardiovascular disease.
Collapse
Affiliation(s)
- Lisa Deng
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), Cardiovascular Research Track, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Luisa Pollmeier
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rebecca Bednarz
- Institute of Experimental Cardiology, Heidelberg University Hospital, Heidelberg, Germany
- DZHK (German Center of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Can Cao
- Institute of Experimental Cardiology, Heidelberg University Hospital, Heidelberg, Germany
- DZHK (German Center of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Patrick Laurette
- Institute of Experimental Cardiology, Heidelberg University Hospital, Heidelberg, Germany
- DZHK (German Center of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Luisa Wirth
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Argen Mamazhakypov
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christine Bode
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental Cardiology, Heidelberg University Hospital, Heidelberg, Germany
- DZHK (German Center of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Interdisciplinary Medical Intensive Care, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
5
|
Savarese G, Lindberg F, Filippatos G, Butler J, Anker SD. Mineralocorticoid receptor overactivation: targeting systemic impact with non-steroidal mineralocorticoid receptor antagonists. Diabetologia 2024; 67:246-262. [PMID: 38127122 PMCID: PMC10789668 DOI: 10.1007/s00125-023-06031-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/13/2023] [Indexed: 12/23/2023]
Abstract
The overactivation of the mineralocorticoid receptor (MR) promotes pathophysiological processes related to multiple physiological systems, including the heart, vasculature, adipose tissue and kidneys. The inhibition of the MR with classical MR antagonists (MRA) has successfully improved outcomes most evidently in heart failure. However, real and perceived risk of side effects and limited tolerability associated with classical MRA have represented barriers to implementing MRA in settings where they have been already proven efficacious (heart failure with reduced ejection fraction) and studying their potential role in settings where they might be beneficial but where risk of safety events is perceived to be higher (renal disease). Novel non-steroidal MRA have distinct properties that might translate into favourable clinical effects and better safety profiles as compared with MRA currently used in clinical practice. Randomised trials have shown benefits of non-steroidal MRA in a range of clinical contexts, including diabetic kidney disease, hypertension and heart failure. This review provides an overview of the literature on the systemic impact of MR overactivation across organ systems. Moreover, we summarise the evidence from preclinical studies and clinical trials that have set the stage for a potential new paradigm of MR antagonism.
Collapse
Affiliation(s)
- Gianluigi Savarese
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
- Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden.
| | - Felix Lindberg
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gerasimos Filippatos
- Department of Cardiology, University Hospital Attikon, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, USA
- Department of Internal Medicine, University of Mississippi, Jackson, MS, USA
| | - Stefan D Anker
- Department of Cardiology (CVK) and Berlin Institute of Health Center for Regenerative Therapies, German Centre for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin, Berlin, Germany.
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland.
| |
Collapse
|
6
|
Crompton M, Skinner LJ, Satchell SC, Butler MJ. Aldosterone: Essential for Life but Damaging to the Vascular Endothelium. Biomolecules 2023; 13:1004. [PMID: 37371584 PMCID: PMC10296074 DOI: 10.3390/biom13061004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
The renin angiotensin aldosterone system is a key regulator of blood pressure. Aldosterone is the final effector of this pathway, acting predominantly via mineralocorticoid receptors. Aldosterone facilitates the conservation of sodium and, with it, water and acts as a powerful stimulus for potassium excretion. However, evidence for the pathological impact of excess mineralocorticoid receptor stimulation is increasing. Here, we discussed how in the heart, hyperaldosteronism is associated with fibrosis, cardiac dysfunction, and maladaptive hypertrophy. In the kidney, aldosterone was shown to cause proteinuria and fibrosis and may contribute to the progression of kidney disease. More recently, studies suggested that aldosterone excess damaged endothelial cells. Here, we reviewed how damage to the endothelial glycocalyx may contribute to this process. The endothelial glycocalyx is a heterogenous, negatively charged layer on the luminal surface of cells. Aldosterone exposure alters this layer. The resulting structural changes reduced endothelial reactivity in response to protective shear stress, altered permeability, and increased immune cell trafficking. Finally, we reviewed current therapeutic strategies for limiting endothelial damage and suggested that preventing glycocalyx remodelling in response to aldosterone exposure may provide a novel strategy, free from the serious adverse effect of hyperkalaemia seen in response to mineralocorticoid blockade.
Collapse
Affiliation(s)
| | | | | | - Matthew J. Butler
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| |
Collapse
|
7
|
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
|
8
|
Liu T, Li YL, Zhou LJ, Sun XN, Wang YL, Du LJ, Liu Y, Zhu H, Chen BY, Sun JY, Liu Y, Xu S, Ye HL, Huang SJ, Wang X, Li B, Duan SZ. Mineralocorticoid Receptor Deficiency in Treg Cells Ameliorates DSS-Induced Colitis in a Gut Microbiota-Dependent Manner. Immunology 2022; 167:94-104. [PMID: 35751882 DOI: 10.1111/imm.13522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/14/2022] [Indexed: 11/29/2022] Open
Abstract
Mineralocorticoid receptor (MR) is a classic nuclear receptor and an effective drug target in the cardiovascular system. The function of MR in immune cells such as macrophages and T cells has been increasingly appreciated. The aim of this study was to investigate the function of Treg MR in the process of inflammatory bowel disease (IBD). We treated Treg MR-deficient (MRflox/flox Foxp3YFP-Cre , KO) mice and control (Foxp3YFP-Cre , WT) mice with dextran sodium sulphate (DSS) to induce colitis and found that the severity of DSS-induced colitis was markedly alleviated in Treg MR-deficient mice, accompanied by reduced production of inflammatory cytokines, and relieved infiltration of monocytes, neutrophils and interferon γ+ T cells in colon lamina propria. Fecal microbiota of mice with colitis was analyzed by 16S rRNA gene sequencing and the composition of gut microbiota was vastly changed in Treg MR-deficient mice. Furthermore, depletion of gut microbiota by antibiotics abolished the protective effects of Treg MR deficiency and resulted in similar severity of DSS-induced colitis in WT and KO mice. Fecal microbiota transplantation from KO mice attenuated DSS-induced colitis characterized by alleviated inflammatory infiltration compared to that from WT mice. Hence, our study demonstrates that Treg MR deficiency protects against DSS-induced colitis by attenuation of colonic inflammatory infiltration. Gut microbiota is both sufficient and necessary for Treg MR deficiency to exert the beneficial effects.
Collapse
Affiliation(s)
- Ting Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yu-Lin Li
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Lu-Jun Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Xue-Nan Sun
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yong-Li Wang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Lin-Juan Du
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yuan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hong Zhu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Bo-Yan Chen
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Jian-Yong Sun
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Shuo Xu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hui-Lin Ye
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Shi-Jia Huang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Xiaoxia Wang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
9
|
Pollard CM, Suster MS, Cora N, Carbone AM, Lymperopoulos A. GRK5 is an essential co-repressor of the cardiac mineralocorticoid receptor and is selectively induced by finerenone. World J Cardiol 2022; 14:220-230. [PMID: 35582468 PMCID: PMC9048278 DOI: 10.4330/wjc.v14.i4.220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/17/2022] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND In the heart, aldosterone (Aldo) binds the mineralocorticoid receptor (MR) to exert damaging, adverse remodeling-promoting effects. We recently showed that G protein-coupled receptor-kinase (GRK)-5 blocks the cardiac MR by directly phosphorylating it, thereby repressing its transcriptional activity. MR antagonist (MRA) drugs block the cardiac MR reducing morbidity and mortality of advanced human heart failure. Non-steroidal MRAs, such as finerenone, may provide better cardio-protection against Aldo than classic, steroidal MRAs, like spironolactone and eplerenone. AIM To investigate potential differences between finerenone and eplerenone at engaging GRK5-dependent cardiac MR phosphorylation and subsequent blockade. METHODS We used H9c2 cardiomyocytes, which endogenously express the MR and GRK5. RESULTS GRK5 phosphorylates the MR in H9c2 cardiomyocytes in response to finerenone but not to eplerenone. Unlike eplerenone, finerenone alone potently and efficiently suppresses cardiac MR transcriptional activity, thus displaying inverse agonism. GRK5 is necessary for finerenone's inverse agonism, since GRK5 genetic deletion renders finerenone incapable of blocking cardiac MR transcriptional activity. Eplerenone alone does not fully suppress cardiac MR basal activity regardless of GRK5 expression levels. Finally, GRK5 is necessary for the anti-apoptotic, anti-oxidative, and anti-fibrotic effects of both finerenone and eplerenone against Aldo, as well as for the higher efficacy and potency of finerenone at blocking Aldo-induced apoptosis, oxidative stress, and fibrosis. CONCLUSION Finerenone, but not eplerenone, induces GRK5-dependent cardiac MR inhibition, which underlies, at least in part, its higher potency and efficacy, compared to eplerenone, as an MRA in the heart. GRK5 acts as a co-repressor of the cardiac MR and is essential for efficient MR antagonism in the myocardium.
Collapse
Affiliation(s)
- 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 33328-2018, United States
| | - Malka S Suster
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, United States
| | - Natalie Cora
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, United States
| | - Alexandra M Carbone
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, United States
| | - 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 33328-2018, United States.
| |
Collapse
|
10
|
Bauersachs J, Lother A. Mineralocorticoid receptor activation and antagonism in cardiovascular disease: cellular and molecular mechanisms. Kidney Int Suppl (2011) 2022; 12:19-26. [DOI: 10.1016/j.kisu.2021.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 02/08/2023] Open
|
11
|
van der Heijden CDCC, Bode M, Riksen NP, Wenzel UO. The role of the mineralocorticoid receptor in immune cells in in cardiovascular disease. Br J Pharmacol 2021; 179:3135-3151. [PMID: 34935128 DOI: 10.1111/bph.15782] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/22/2021] [Accepted: 11/22/2021] [Indexed: 11/29/2022] Open
Abstract
Chronic low-grade inflammation and immune cell activation are important mechanisms in the pathophysiology of cardiovascular disease (CVD). Therefore, targeted immunosuppression is a promising novel therapy to lower cardiovascular risk. In this review, we identify the mineralocorticoid receptor (MR) on immune cells as a potential target to modulate inflammation. The MR is present in almost all cells of the cardiovascular system, including immune cells. Activation of the MR in innate and adaptive immune cells induces inflammation which can contribute to CVD, by inducing endothelial dysfunction and hypertension. Moreover, it accelerates atherosclerotic plaque formation and destabilization and impairs tissue regeneration after ischemic events. Identifying the molecular targets for these non-renal actions of the MR provide promising novel cardiovascular drug targets for mineralocorticoid receptor antagonists (MRAs), which are currently mainly applied in hypertension and heart failure.
Collapse
Affiliation(s)
| | - Marlies Bode
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, GA, Nijmegen, The Netherlands.,Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, GA, The Netherlands
| | - Ulrich O Wenzel
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
12
|
Pitt B, Filippatos G, Agarwal R, Anker SD, Bakris GL, Rossing P, Joseph A, Kolkhof P, Nowack C, Schloemer P, Ruilope LM. Cardiovascular Events with Finerenone in Kidney Disease and Type 2 Diabetes. N Engl J Med 2021; 385:2252-2263. [PMID: 34449181 DOI: 10.1056/nejmoa2110956] [Citation(s) in RCA: 783] [Impact Index Per Article: 195.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Finerenone, a selective nonsteroidal mineralocorticoid receptor antagonist, has favorable effects on cardiorenal outcomes in patients with predominantly stage 3 or 4 chronic kidney disease (CKD) with severely elevated albuminuria and type 2 diabetes. The use of finerenone in patients with type 2 diabetes and a wider range of CKD is unclear. METHODS In this double-blind trial, we randomly assigned patients with CKD and type 2 diabetes to receive finerenone or placebo. Eligible patients had a urinary albumin-to-creatinine ratio (with albumin measured in milligrams and creatinine measured in grams) of 30 to less than 300 and an estimated glomerular filtration rate (eGFR) of 25 to 90 ml per minute per 1.73 m2 of body-surface area (stage 2 to 4 CKD) or a urinary albumin-to-creatinine ratio of 300 to 5000 and an eGFR of at least 60 ml per minute per 1.73 m2 (stage 1 or 2 CKD). Patients were treated with renin-angiotensin system blockade that had been adjusted before randomization to the maximum dose on the manufacturer's label that did not cause unacceptable side effects. The primary outcome, assessed in a time-to-event analysis, was a composite of death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or hospitalization for heart failure. The first secondary outcome was a composite of kidney failure, a sustained decrease from baseline of at least 40% in the eGFR, or death from renal causes. Safety was assessed as investigator-reported adverse events. RESULTS A total of 7437 patients underwent randomization. Among the patients included in the analysis, during a median follow-up of 3.4 years, a primary outcome event occurred in 458 of 3686 patients (12.4%) in the finerenone group and in 519 of 3666 (14.2%) in the placebo group (hazard ratio, 0.87; 95% confidence interval [CI], 0.76 to 0.98; P = 0.03), with the benefit driven primarily by a lower incidence of hospitalization for heart failure (hazard ratio, 0.71; 95% CI, 0.56 to 0.90). The secondary composite outcome occurred in 350 patients (9.5%) in the finerenone group and in 395 (10.8%) in the placebo group (hazard ratio, 0.87; 95% CI, 0.76 to 1.01). The overall frequency of adverse events did not differ substantially between groups. The incidence of hyperkalemia-related discontinuation of the trial regimen was higher with finerenone (1.2%) than with placebo (0.4%). CONCLUSIONS Among patients with type 2 diabetes and stage 2 to 4 CKD with moderately elevated albuminuria or stage 1 or 2 CKD with severely elevated albuminuria, finerenone therapy improved cardiovascular outcomes as compared with placebo. (Funded by Bayer; FIGARO-DKD ClinicalTrials.gov number, NCT02545049.).
Collapse
Affiliation(s)
- Bertram Pitt
- From the Department of Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); National and Kapodistrian University of Athens, School of Medicine, Department of Cardiology, Attikon University Hospital, Athens (G.F.); the Richard L. Roudebush Veterans Affairs Medical Center and Indiana University, Indianapolis (R.A.); the Department of Cardiology and Berlin Institute of Health Center for Regenerative Therapies, German Center for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin (S.D.A.), and Research and Development, Cardiology and Nephrology Clinical Development (A.J.) and Statistics and Data Insights (P.S.), Bayer, Berlin, and Research and Development, Preclinical Research Cardiovascular (P.K.) and Clinical Development Operations (C.N.), Bayer, Wuppertal - all in Germany; the Department of Medicine, University of Chicago Medicine, Chicago (G.L.B.); Steno Diabetes Center Copenhagen, Gentofte, and the Department of Clinical Medicine, University of Copenhagen, Copenhagen - both in Denmark (P.R.); and the Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research i+12, Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Hospital Universitario 12 de Octubre, and the Faculty of Sport Sciences, European University of Madrid - all in Madrid (L.M.R.)
| | - Gerasimos Filippatos
- From the Department of Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); National and Kapodistrian University of Athens, School of Medicine, Department of Cardiology, Attikon University Hospital, Athens (G.F.); the Richard L. Roudebush Veterans Affairs Medical Center and Indiana University, Indianapolis (R.A.); the Department of Cardiology and Berlin Institute of Health Center for Regenerative Therapies, German Center for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin (S.D.A.), and Research and Development, Cardiology and Nephrology Clinical Development (A.J.) and Statistics and Data Insights (P.S.), Bayer, Berlin, and Research and Development, Preclinical Research Cardiovascular (P.K.) and Clinical Development Operations (C.N.), Bayer, Wuppertal - all in Germany; the Department of Medicine, University of Chicago Medicine, Chicago (G.L.B.); Steno Diabetes Center Copenhagen, Gentofte, and the Department of Clinical Medicine, University of Copenhagen, Copenhagen - both in Denmark (P.R.); and the Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research i+12, Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Hospital Universitario 12 de Octubre, and the Faculty of Sport Sciences, European University of Madrid - all in Madrid (L.M.R.)
| | - Rajiv Agarwal
- From the Department of Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); National and Kapodistrian University of Athens, School of Medicine, Department of Cardiology, Attikon University Hospital, Athens (G.F.); the Richard L. Roudebush Veterans Affairs Medical Center and Indiana University, Indianapolis (R.A.); the Department of Cardiology and Berlin Institute of Health Center for Regenerative Therapies, German Center for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin (S.D.A.), and Research and Development, Cardiology and Nephrology Clinical Development (A.J.) and Statistics and Data Insights (P.S.), Bayer, Berlin, and Research and Development, Preclinical Research Cardiovascular (P.K.) and Clinical Development Operations (C.N.), Bayer, Wuppertal - all in Germany; the Department of Medicine, University of Chicago Medicine, Chicago (G.L.B.); Steno Diabetes Center Copenhagen, Gentofte, and the Department of Clinical Medicine, University of Copenhagen, Copenhagen - both in Denmark (P.R.); and the Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research i+12, Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Hospital Universitario 12 de Octubre, and the Faculty of Sport Sciences, European University of Madrid - all in Madrid (L.M.R.)
| | - Stefan D Anker
- From the Department of Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); National and Kapodistrian University of Athens, School of Medicine, Department of Cardiology, Attikon University Hospital, Athens (G.F.); the Richard L. Roudebush Veterans Affairs Medical Center and Indiana University, Indianapolis (R.A.); the Department of Cardiology and Berlin Institute of Health Center for Regenerative Therapies, German Center for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin (S.D.A.), and Research and Development, Cardiology and Nephrology Clinical Development (A.J.) and Statistics and Data Insights (P.S.), Bayer, Berlin, and Research and Development, Preclinical Research Cardiovascular (P.K.) and Clinical Development Operations (C.N.), Bayer, Wuppertal - all in Germany; the Department of Medicine, University of Chicago Medicine, Chicago (G.L.B.); Steno Diabetes Center Copenhagen, Gentofte, and the Department of Clinical Medicine, University of Copenhagen, Copenhagen - both in Denmark (P.R.); and the Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research i+12, Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Hospital Universitario 12 de Octubre, and the Faculty of Sport Sciences, European University of Madrid - all in Madrid (L.M.R.)
| | - George L Bakris
- From the Department of Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); National and Kapodistrian University of Athens, School of Medicine, Department of Cardiology, Attikon University Hospital, Athens (G.F.); the Richard L. Roudebush Veterans Affairs Medical Center and Indiana University, Indianapolis (R.A.); the Department of Cardiology and Berlin Institute of Health Center for Regenerative Therapies, German Center for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin (S.D.A.), and Research and Development, Cardiology and Nephrology Clinical Development (A.J.) and Statistics and Data Insights (P.S.), Bayer, Berlin, and Research and Development, Preclinical Research Cardiovascular (P.K.) and Clinical Development Operations (C.N.), Bayer, Wuppertal - all in Germany; the Department of Medicine, University of Chicago Medicine, Chicago (G.L.B.); Steno Diabetes Center Copenhagen, Gentofte, and the Department of Clinical Medicine, University of Copenhagen, Copenhagen - both in Denmark (P.R.); and the Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research i+12, Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Hospital Universitario 12 de Octubre, and the Faculty of Sport Sciences, European University of Madrid - all in Madrid (L.M.R.)
| | - Peter Rossing
- From the Department of Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); National and Kapodistrian University of Athens, School of Medicine, Department of Cardiology, Attikon University Hospital, Athens (G.F.); the Richard L. Roudebush Veterans Affairs Medical Center and Indiana University, Indianapolis (R.A.); the Department of Cardiology and Berlin Institute of Health Center for Regenerative Therapies, German Center for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin (S.D.A.), and Research and Development, Cardiology and Nephrology Clinical Development (A.J.) and Statistics and Data Insights (P.S.), Bayer, Berlin, and Research and Development, Preclinical Research Cardiovascular (P.K.) and Clinical Development Operations (C.N.), Bayer, Wuppertal - all in Germany; the Department of Medicine, University of Chicago Medicine, Chicago (G.L.B.); Steno Diabetes Center Copenhagen, Gentofte, and the Department of Clinical Medicine, University of Copenhagen, Copenhagen - both in Denmark (P.R.); and the Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research i+12, Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Hospital Universitario 12 de Octubre, and the Faculty of Sport Sciences, European University of Madrid - all in Madrid (L.M.R.)
| | - Amer Joseph
- From the Department of Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); National and Kapodistrian University of Athens, School of Medicine, Department of Cardiology, Attikon University Hospital, Athens (G.F.); the Richard L. Roudebush Veterans Affairs Medical Center and Indiana University, Indianapolis (R.A.); the Department of Cardiology and Berlin Institute of Health Center for Regenerative Therapies, German Center for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin (S.D.A.), and Research and Development, Cardiology and Nephrology Clinical Development (A.J.) and Statistics and Data Insights (P.S.), Bayer, Berlin, and Research and Development, Preclinical Research Cardiovascular (P.K.) and Clinical Development Operations (C.N.), Bayer, Wuppertal - all in Germany; the Department of Medicine, University of Chicago Medicine, Chicago (G.L.B.); Steno Diabetes Center Copenhagen, Gentofte, and the Department of Clinical Medicine, University of Copenhagen, Copenhagen - both in Denmark (P.R.); and the Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research i+12, Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Hospital Universitario 12 de Octubre, and the Faculty of Sport Sciences, European University of Madrid - all in Madrid (L.M.R.)
| | - Peter Kolkhof
- From the Department of Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); National and Kapodistrian University of Athens, School of Medicine, Department of Cardiology, Attikon University Hospital, Athens (G.F.); the Richard L. Roudebush Veterans Affairs Medical Center and Indiana University, Indianapolis (R.A.); the Department of Cardiology and Berlin Institute of Health Center for Regenerative Therapies, German Center for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin (S.D.A.), and Research and Development, Cardiology and Nephrology Clinical Development (A.J.) and Statistics and Data Insights (P.S.), Bayer, Berlin, and Research and Development, Preclinical Research Cardiovascular (P.K.) and Clinical Development Operations (C.N.), Bayer, Wuppertal - all in Germany; the Department of Medicine, University of Chicago Medicine, Chicago (G.L.B.); Steno Diabetes Center Copenhagen, Gentofte, and the Department of Clinical Medicine, University of Copenhagen, Copenhagen - both in Denmark (P.R.); and the Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research i+12, Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Hospital Universitario 12 de Octubre, and the Faculty of Sport Sciences, European University of Madrid - all in Madrid (L.M.R.)
| | - Christina Nowack
- From the Department of Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); National and Kapodistrian University of Athens, School of Medicine, Department of Cardiology, Attikon University Hospital, Athens (G.F.); the Richard L. Roudebush Veterans Affairs Medical Center and Indiana University, Indianapolis (R.A.); the Department of Cardiology and Berlin Institute of Health Center for Regenerative Therapies, German Center for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin (S.D.A.), and Research and Development, Cardiology and Nephrology Clinical Development (A.J.) and Statistics and Data Insights (P.S.), Bayer, Berlin, and Research and Development, Preclinical Research Cardiovascular (P.K.) and Clinical Development Operations (C.N.), Bayer, Wuppertal - all in Germany; the Department of Medicine, University of Chicago Medicine, Chicago (G.L.B.); Steno Diabetes Center Copenhagen, Gentofte, and the Department of Clinical Medicine, University of Copenhagen, Copenhagen - both in Denmark (P.R.); and the Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research i+12, Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Hospital Universitario 12 de Octubre, and the Faculty of Sport Sciences, European University of Madrid - all in Madrid (L.M.R.)
| | - Patrick Schloemer
- From the Department of Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); National and Kapodistrian University of Athens, School of Medicine, Department of Cardiology, Attikon University Hospital, Athens (G.F.); the Richard L. Roudebush Veterans Affairs Medical Center and Indiana University, Indianapolis (R.A.); the Department of Cardiology and Berlin Institute of Health Center for Regenerative Therapies, German Center for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin (S.D.A.), and Research and Development, Cardiology and Nephrology Clinical Development (A.J.) and Statistics and Data Insights (P.S.), Bayer, Berlin, and Research and Development, Preclinical Research Cardiovascular (P.K.) and Clinical Development Operations (C.N.), Bayer, Wuppertal - all in Germany; the Department of Medicine, University of Chicago Medicine, Chicago (G.L.B.); Steno Diabetes Center Copenhagen, Gentofte, and the Department of Clinical Medicine, University of Copenhagen, Copenhagen - both in Denmark (P.R.); and the Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research i+12, Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Hospital Universitario 12 de Octubre, and the Faculty of Sport Sciences, European University of Madrid - all in Madrid (L.M.R.)
| | - Luis M Ruilope
- From the Department of Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); National and Kapodistrian University of Athens, School of Medicine, Department of Cardiology, Attikon University Hospital, Athens (G.F.); the Richard L. Roudebush Veterans Affairs Medical Center and Indiana University, Indianapolis (R.A.); the Department of Cardiology and Berlin Institute of Health Center for Regenerative Therapies, German Center for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin (S.D.A.), and Research and Development, Cardiology and Nephrology Clinical Development (A.J.) and Statistics and Data Insights (P.S.), Bayer, Berlin, and Research and Development, Preclinical Research Cardiovascular (P.K.) and Clinical Development Operations (C.N.), Bayer, Wuppertal - all in Germany; the Department of Medicine, University of Chicago Medicine, Chicago (G.L.B.); Steno Diabetes Center Copenhagen, Gentofte, and the Department of Clinical Medicine, University of Copenhagen, Copenhagen - both in Denmark (P.R.); and the Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research i+12, Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Hospital Universitario 12 de Octubre, and the Faculty of Sport Sciences, European University of Madrid - all in Madrid (L.M.R.)
| |
Collapse
|
13
|
Ruhs S, Griesler B, Huebschmann R, Stroedecke K, Straetz N, Ihling C, Sinz A, Masch A, Schutkowski M, Gekle M, Grossmann C. Modulation of transcriptional mineralocorticoid receptor activity by casein kinase 1. FASEB J 2021; 36:e22059. [PMID: 34847273 DOI: 10.1096/fj.202100977rr] [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: 06/14/2021] [Revised: 10/16/2021] [Accepted: 11/08/2021] [Indexed: 11/11/2022]
Abstract
The mineralocorticoid receptor (MR) with its ligand aldosterone (aldo) physiologically regulates electrolyte homeostasis and blood pressure but it can also lead to pathophysiological effects in the cardiovascular system. Previous results show that posttranslational modifications (PTM) can influence MR signaling and function. Based on in silico and in vitro data, casein kinase 1 (CK1) was predicted as a candidate for MR phosphorylation. To gain a deeper mechanistic insight into MR activation, we investigated the influence of CK1 on MR function in HEK cells. Co-immunoprecipitation experiments indicated that the MR is located in a protein-protein complex with CK1α and CK1ε. Reporter gene assays with pharmacological inhibitors and MR constructs demonstrated that especially CK1ε acts as a positive modulator of GRE activity via the C-terminal MR domains CDEF. CK1 enhanced the binding affinity of aldosterone to the MR, facilitated nuclear translocation and DNA interaction of the MR, and led to expression changes of pathophysiologically relevant genes like Per-1 and Phlda1. By peptide microarray and site-directed mutagenesis experiments, we identified the highly conserved T800 as a direct CK1 phosphorylation site of the MR, which modulates the nuclear import and genomic activity of the receptor. Direct phosphorylation of the MR was unable to fully account for all of the CK1 effects on MR signaling, suggesting additional phosphorylation of MR co-regulators. By LC/MS/MS, we identified the MR-associated proteins NOLC1 and TCOF1 as candidates for such CK1-regulated co-factors. Overall, we found that CK1 acts as a co-activator of MR GRE activity through direct and indirect phosphorylation, which accelerates cytosolic-nuclear trafficking, facilitates nuclear accumulation and DNA binding of the MR, and increases the expression of pathologically relevant MR-target genes.
Collapse
Affiliation(s)
- Stefanie Ruhs
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.,Department of Anesthesiology and Surgical Intensive Care, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Bruno Griesler
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Ralf Huebschmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Katharina Stroedecke
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Nicole Straetz
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Christian Ihling
- Department of Pharmaceutical Chemistry & Bioanalytics, Center for Structural Mass Spectrometry, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Andrea Sinz
- Department of Pharmaceutical Chemistry & Bioanalytics, Center for Structural Mass Spectrometry, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Antonia Masch
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Martin Luther University of Halle-Wittenberg, Halle (Saale), Germany
| | - Mike Schutkowski
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Martin Luther University of Halle-Wittenberg, Halle (Saale), Germany
| | - Michael Gekle
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Claudia Grossmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
14
|
Mamazhakypov A, Hein L, Lother A. Mineralocorticoid receptors in pulmonary hypertension and right heart failure: From molecular biology to therapeutic targeting. Pharmacol Ther 2021; 231:107987. [PMID: 34480966 DOI: 10.1016/j.pharmthera.2021.107987] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/21/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022]
Abstract
Pulmonary hypertension (PH) is a devastating condition characterized by pulmonary vascular remodelling, leading to progressive increase in pulmonary artery pressure and subsequent right ventricular failure. Aldosterone and the mineralocorticoid receptor (MR), a nuclear transcription factor, are key drivers of cardiovascular disease and MR antagonists are well-established in heart failure. Now, a growing body of evidence points at a detrimental role of MR in PH. Pharmacological MR blockade attenuated PH and prevented RV failure in experimental models. Mouse models with cell selective MR deletion suggest that this effect is mediated by MR in endothelial cells. While the evidence from experimental studies appears convincing, the available clinical data on MR antagonist use in patients with PH is more controversial. Integrated analysis of clinical data together with MR-dependent molecular alterations may provide insights why some patients respond to MRA treatment while others do not. Potential ways to identify MRA 'responders' include the analysis of underlying PH causes, stage of disease, or sex, as well as new biomarkers.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany; Heart Center Freiburg University, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Germany.
| |
Collapse
|
15
|
Kolkhof P, Joseph A, Kintscher U. Nonsteroidal mineralocorticoid receptor antagonism for cardiovascular and renal disorders - New perspectives for combination therapy. Pharmacol Res 2021; 172:105859. [PMID: 34461222 DOI: 10.1016/j.phrs.2021.105859] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/16/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023]
Abstract
During the recent 30 years, there has been a dramatic increase in knowledge about the role of aldosterone and the mineralocorticoid receptor (MR) in the pathophysiology of cardiovascular (CV) and kidney diseases. The scientific perspective on the aldosterone/MR ensemble extended from a previously renal epithelial-centered focus on sodium-potassium exchange to a broader view as systemic modulators of extracellular matrix, inflammation and fibrosis. Spironolactone was launched as the first antagonist of aldosterone 27 years before the MR was cloned. It was classified as a potassium-sparing diuretic, based on its initial clinical characterization as a diuretic and its preferred activity to compensate for the potassium loss induced by loop diuretics when used in combination. The second steroidal MR antagonist was eplerenone which was discovered at a time when the role of aldosterone and MR in cardiac fibrosis was rediscovered. The constraint of developing potentially life-threatening hyperkalaemia when used in combination with other inhibitors of the renin-angiotensin-system (RAS) in patients with reduced kidney function initiated extensive research and development activities with the goal to identify novel nonsteroidal MR antagonists with an improved benefit-risk ratio. Here we summarize major current clinical trials with MRAs in different CV and renal diseases. Addition of the nonsteroidal MRA finerenone to optimal RAS blockade recently reduced CV and kidney outcomes in two large phase III trials in patients with chronic kidney disease (CKD) and type 2 diabetes (T2D). We provide an outlook on further opportunities for combination therapy of nonsteroidal MRA finerenone with RAS inhibitors and sodium-glucose cotransporter-2 inhibitors (SGLT2i).
Collapse
Affiliation(s)
- Peter Kolkhof
- Cardiovascular Research, Research and Early Development, R&D Pharmaceuticals, Bayer AG, Wuppertal, Germany.
| | - Amer Joseph
- Cardiology and Nephrology, Clinical Development, R&D Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Ulrich Kintscher
- Charite - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal Research Center, 10115 Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| |
Collapse
|
16
|
Kowalski J, Deng L, Suennen C, Koca D, Meral D, Bode C, Hein L, Lother A. Eplerenone Improves Pulmonary Vascular Remodeling and Hypertension by Inhibition of the Mineralocorticoid Receptor in Endothelial Cells. Hypertension 2021; 78:456-465. [PMID: 33966455 DOI: 10.1161/hypertensionaha.120.16196] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jessica Kowalski
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany
| | - Lisa Deng
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany
| | - Chiara Suennen
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany
| | - Duygu Koca
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany
| | - David Meral
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), Cardiovascular Research Track (D.M.), University of Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, Faculty of Medicine, Heart Center Freiburg University (C.B., A.L.), University of Freiburg, Germany
| | - Lutz Hein
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies (L.H.), University of Freiburg, Germany
| | - Achim Lother
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine (J.K., L.D., C.S., D.K., D.M., L.H., A.L.), University of Freiburg, Germany.,Department of Cardiology and Angiology I, Faculty of Medicine, Heart Center Freiburg University (C.B., A.L.), University of Freiburg, Germany
| |
Collapse
|
17
|
Ito J, Minemura T, Wälchli S, Niimi T, Fujihara Y, Kuroda S, Takimoto K, Maturana AD. Id2 Represses Aldosterone-Stimulated Cardiac T-Type Calcium Channels Expression. Int J Mol Sci 2021; 22:3561. [PMID: 33808082 PMCID: PMC8037527 DOI: 10.3390/ijms22073561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/16/2022] Open
Abstract
Aldosterone excess is a cardiovascular risk factor. Aldosterone can directly stimulate an electrical remodeling of cardiomyocytes leading to cardiac arrhythmia and hypertrophy. L-type and T-type voltage-gated calcium (Ca2+) channels expression are increased by aldosterone in cardiomyocytes. To further understand the regulation of these channels expression, we studied the role of a transcriptional repressor, the inhibitor of differentiation/DNA binding protein 2 (Id2). We found that aldosterone inhibited the expression of Id2 in neonatal rat cardiomyocytes and in the heart of adult mice. When Id2 was overexpressed in cardiomyocytes, we observed a reduction in the spontaneous action potentials rate and an arrest in aldosterone-stimulated rate increase. Accordingly, Id2 siRNA knockdown increased this rate. We also observed that CaV1.2 (L-type Ca2+ channel) or CaV3.1, and CaV3.2 (T-type Ca2+ channels) mRNA expression levels and Ca2+ currents were affected by Id2 presence. These observations were further corroborated in a heart specific Id2- transgenic mice. Taken together, our results suggest that Id2 functions as a transcriptional repressor for L- and T-type Ca2+ channels, particularly CaV3.1, in cardiomyocytes and its expression is controlled by aldosterone. We propose that Id2 might contributes to a protective mechanism in cardiomyocytes preventing the presence of channels associated with a pathological state.
Collapse
Affiliation(s)
- Jumpei Ito
- Laboratory of Animal Cell Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan; (J.I.); (T.M.); (T.N.)
| | - Tomomi Minemura
- Laboratory of Animal Cell Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan; (J.I.); (T.M.); (T.N.)
| | - Sébastien Wälchli
- Translational Research Unit, Section for Cellular Therapy, Oslo University Hospital, 0379 Oslo, Norway;
| | - Tomoaki Niimi
- Laboratory of Animal Cell Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan; (J.I.); (T.M.); (T.N.)
| | - Yoshitaka Fujihara
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka 565-0871, Japan;
| | - Shun’ichi Kuroda
- Institute for Scientific and Industrial Researches, Osaka University, Osaka 567-0047, Japan;
| | - Koichi Takimoto
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka 940-2188, Japan;
| | - Andrés D. Maturana
- Laboratory of Animal Cell Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan; (J.I.); (T.M.); (T.N.)
| |
Collapse
|
18
|
Affiliation(s)
- Achim Lother
- From the Department of Cardiology and Angiology I, Faculty of Medicine, Heart Center Freiburg University, University of Freiburg, Germany (A.L.)
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany (A.L.)
| |
Collapse
|
19
|
Feldman RD, Hegele RA. Primary Aldosteronism in Hypertension: More Than a Factoid. Can J Cardiol 2020; 37:196-198. [PMID: 32628977 DOI: 10.1016/j.cjca.2020.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 11/19/2022] Open
Affiliation(s)
- Ross D Feldman
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada; Cardiac Sciences Program, Winnipeg Regional Health Authority, Winnipeg, Manitoba, Canada.
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
20
|
Zheng W, Ocorr K, Tatar M. Extracellular matrix induced by steroids and aging through a G-protein-coupled receptor in a Drosophila model of renal fibrosis. Dis Model Mech 2020; 13:dmm041301. [PMID: 32461236 PMCID: PMC7328168 DOI: 10.1242/dmm.041301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 04/17/2020] [Indexed: 12/20/2022] Open
Abstract
Aldosterone is produced by the mammalian adrenal cortex to modulate blood pressure and fluid balance; however, excessive, prolonged aldosterone promotes fibrosis and kidney failure. How aldosterone triggers disease may involve actions independent of its canonical mineralocorticoid receptor. Here, we present a Drosophila model of renal pathology caused by excess extracellular matrix formation, stimulated by exogenous aldosterone and by insect ecdysone. Chronic administration of aldosterone or ecdysone induces expression and accumulation of collagen-like Pericardin in adult nephrocytes - podocyte-like cells that filter circulating hemolymph. Excess Pericardin deposition disrupts nephrocyte (glomerular) filtration and causes proteinuria in Drosophila, hallmarks of mammalian kidney failure. Steroid-induced Pericardin production arises from cardiomyocytes associated with nephrocytes, potentially reflecting an analogous role of mammalian myofibroblasts in fibrotic disease. Remarkably, the canonical ecdysteroid nuclear hormone receptor, Ecdysone receptor (EcR), is not required for aldosterone or ecdysone to stimulate Pericardin production or associated renal pathology. Instead, these hormones require a cardiomyocyte-associated G-protein-coupled receptor, Dopamine-EcR (DopEcR), a membrane-associated receptor previously characterized in the fly brain to affect behavior. DopEcR in the brain is known to affect behavior through interactions with the Drosophila Epidermal growth factor receptor (Egfr), referred to as dEGFR. Here, we find that the steroids ecdysone and aldosterone require dEGFR in cardiomyocytes to induce fibrosis of the cardiac-renal system. In addition, endogenous ecdysone that becomes elevated with age is found to foster age-associated fibrosis, and to require both cardiomyocyte DopEcR and dEGFR. This Drosophila renal disease model reveals a novel signaling pathway through which steroids may modulate mammalian fibrosis through potential orthologs of DopEcR.
Collapse
Affiliation(s)
- Wenjing Zheng
- Department of Ecology and Evolutionary Biology, Division of Biology and Medicine, Brown University, Providence RI 02912, USA
| | - Karen Ocorr
- Development, Aging and Regeneration Program, SBP Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Marc Tatar
- Department of Ecology and Evolutionary Biology, Division of Biology and Medicine, Brown University, Providence RI 02912, USA
| |
Collapse
|
21
|
Yan C, Quan XJ, Feng YM. Nanomedicine for Gene Delivery for the Treatment of Cardiovascular Diseases. Curr Gene Ther 2020; 19:20-30. [PMID: 30280665 PMCID: PMC6751340 DOI: 10.2174/1566523218666181003125308] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/21/2018] [Accepted: 09/13/2018] [Indexed: 12/13/2022]
Abstract
Background: Myocardial infarction (MI) is the most severe ischemic heart disease and di-rectly leads to heart failure till death. Target molecules have been identified in the event of MI including increasing angiogenesis, promoting cardiomyocyte survival, improving heart function and restraining inflammation and myocyte activation and subsequent fibrosis. All of which are substantial in cardiomy-ocyte protection and preservation of cardiac function. Methodology: To modulate target molecule expression, virus and non-virus-mediated gene transfer have been investigated. Despite successful in animal models of MI, virus-mediated gene transfer is hampered by poor targeting efficiency, low packaging capacity for large DNA sequences, immunogenicity induced by virus and random integration into the human genome. Discussion: Nanoparticles could be synthesized and equipped on purpose for large-scale production. They are relatively small in size and do not incorporate into the genome. They could carry DNA and drug within the same transfer. All of these properties make them an alternative strategy for gene transfer. In the review, we first introduce the pathological progression of MI. After concise discussion on the current status of virus-mediated gene therapy in treating MI, we overview the history and development of nanoparticle-based gene delivery system. We point out the limitations and future perspective in the field of nanoparticle vehicle. Conclusion: Ultimately, we hope that this review could help to better understand how far we are with nanoparticle-facilitated gene transfer strategy and what obstacles we need to solve for utilization of na-nomedicine in the treatment of MI.
Collapse
Affiliation(s)
- Cen Yan
- Beijing Key Laboratory of Diabetes Prevention and Research, Endocrinology Center, Lu He Hospital, Capital Medical University, Beijing 101149, China
| | - Xiao-Jiang Quan
- Laboratory of Brain Development, Institut du Cerveau et de la Moelle Epiniere- ICM, Hospital Pitie-Salpetriere, 75013 Paris, France
| | - Ying-Mei Feng
- Beijing Key Laboratory of Diabetes Prevention and Research, Endocrinology Center, Lu He Hospital, Capital Medical University, Beijing 101149, China
| |
Collapse
|
22
|
Maning J, McCrink KA, Pollard CM, Desimine VL, Ghandour J, Perez A, Cora N, Ferraino KE, Parker BM, Brill AR, Aukszi B, Lymperopoulos A. Antagonistic Roles of GRK2 and GRK5 in Cardiac Aldosterone Signaling Reveal GRK5-Mediated Cardioprotection via Mineralocorticoid Receptor Inhibition. Int J Mol Sci 2020; 21:2868. [PMID: 32326036 PMCID: PMC7215681 DOI: 10.3390/ijms21082868] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/14/2020] [Accepted: 04/17/2020] [Indexed: 12/13/2022] Open
Abstract
Aldosterone (Aldo), when overproduced, is a cardiotoxic hormone underlying heart failure and hypertension. Aldo exerts damaging effects via the mineralocorticoid receptor (MR) but also activates the antiapoptotic G protein-coupled estrogen receptor (GPER) in the heart. G protein-coupled receptor (GPCR)-kinase (GRK)-2 and -5 are the most abundant cardiac GRKs and phosphorylate GPCRs as well as non-GPCR substrates. Herein, we investigated whether they phosphorylate and regulate cardiac MR and GPER. To this end, we used the cardiomyocyte cell line H9c2 and adult rat ventricular myocytes (ARVMs), in which we manipulated GRK5 protein levels via clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 and GRK2 activity via pharmacological inhibition. We report that GRK5 phosphorylates and inhibits the cardiac MR whereas GRK2 phosphorylates and desensitizes GPER. In H9c2 cardiomyocytes, GRK5 interacts with and phosphorylates the MR upon β2-adrenergic receptor (AR) activation. In contrast, GRK2 opposes agonist-activated GPER signaling. Importantly, GRK5-dependent MR phosphorylation of the MR inhibits transcriptional activity, since aldosterone-induced gene transcription is markedly suppressed in GRK5-overexpressing cardiomyocytes. Conversely, GRK5 gene deletion augments cardiac MR transcriptional activity. β2AR-stimulated GRK5 phosphorylates and inhibits the MR also in ARVMs. Additionally, GRK5 is necessary for the protective effects of the MR antagonist drug eplerenone against Aldo-induced apoptosis and oxidative stress in ARVMs. In conclusion, GRK5 blocks the cardiotoxic MR-dependent effects of Aldo in the heart, whereas GRK2 may hinder beneficial effects of Aldo through GPER. Thus, cardiac GRK5 stimulation (e.g., via β2AR activation) might be of therapeutic value for heart disease treatment via boosting the efficacy of MR antagonists against Aldo-mediated cardiac injury.
Collapse
Affiliation(s)
- Jennifer Maning
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.M.); (K.A.M.); (C.M.P.); (V.L.D.); (J.G.); (A.P.); (N.C.); (K.E.F.); (B.M.P.); (A.R.B.)
| | - Katie A. McCrink
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.M.); (K.A.M.); (C.M.P.); (V.L.D.); (J.G.); (A.P.); (N.C.); (K.E.F.); (B.M.P.); (A.R.B.)
| | - Celina M. Pollard
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.M.); (K.A.M.); (C.M.P.); (V.L.D.); (J.G.); (A.P.); (N.C.); (K.E.F.); (B.M.P.); (A.R.B.)
| | - Victoria L. Desimine
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.M.); (K.A.M.); (C.M.P.); (V.L.D.); (J.G.); (A.P.); (N.C.); (K.E.F.); (B.M.P.); (A.R.B.)
| | - Jennifer Ghandour
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.M.); (K.A.M.); (C.M.P.); (V.L.D.); (J.G.); (A.P.); (N.C.); (K.E.F.); (B.M.P.); (A.R.B.)
| | - Arianna Perez
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.M.); (K.A.M.); (C.M.P.); (V.L.D.); (J.G.); (A.P.); (N.C.); (K.E.F.); (B.M.P.); (A.R.B.)
| | - Natalie Cora
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.M.); (K.A.M.); (C.M.P.); (V.L.D.); (J.G.); (A.P.); (N.C.); (K.E.F.); (B.M.P.); (A.R.B.)
| | - Krysten E. Ferraino
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.M.); (K.A.M.); (C.M.P.); (V.L.D.); (J.G.); (A.P.); (N.C.); (K.E.F.); (B.M.P.); (A.R.B.)
| | - Barbara M. Parker
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.M.); (K.A.M.); (C.M.P.); (V.L.D.); (J.G.); (A.P.); (N.C.); (K.E.F.); (B.M.P.); (A.R.B.)
| | - Ava R. Brill
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.M.); (K.A.M.); (C.M.P.); (V.L.D.); (J.G.); (A.P.); (N.C.); (K.E.F.); (B.M.P.); (A.R.B.)
| | - Beatrix Aukszi
- Department of Chemistry and Physics, Halmos College of Natural Sciences and Oceanography, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| | - Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.M.); (K.A.M.); (C.M.P.); (V.L.D.); (J.G.); (A.P.); (N.C.); (K.E.F.); (B.M.P.); (A.R.B.)
| |
Collapse
|
23
|
Lother A, Bergemann S, Kowalski J, Huck M, Gilsbach R, Bode C, Hein L. Inhibition of the cardiac myocyte mineralocorticoid receptor ameliorates doxorubicin-induced cardiotoxicity. Cardiovasc Res 2019; 114:282-290. [PMID: 28430882 DOI: 10.1093/cvr/cvx078] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 04/13/2017] [Indexed: 11/13/2022] Open
Abstract
Aim Anthracyclines such as doxorubicin are widely used in cancer therapy but their use is limited by cardiotoxicity. Up to date there is no established strategy for the prevention of anthracyclin-induced heart failure. In this study, we evaluated the role of the cardiac myocyte mineralocorticoid receptor (MR) during doxorubicin-induced cardiotoxicity. Methods and results A single high-dose or repetitive low-dose doxorubicin administration lead to markedly reduced left ventricular function in mice. Treatment with the MR antagonist eplerenone prevented doxorubicin-induced left ventricular dysfunction. In order to identify the cell types and molecular mechanisms involved in this beneficial effect we used a mouse model with cell type-specific MR deletion in cardiac myocytes. Cardiac myocyte MR deletion largely reproduced the effect of pharmacological MR inhibition on doxorubicin-induced cardiotoxicity. RNAseq from isolated cardiac myocytes revealed a repressive effect of doxorubicin on gene expression which was prevented by MR deletion. Conclusions We show here that (i) eplerenone prevents doxorubicin-induced left ventricular dysfunction in mice, and (ii) this beneficial effect is related to inhibition of MR in cardiac myocytes. Together with present clinical trial data our findings suggest that MR antagonism may be appropriate for the prevention of doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany.,Heart Center, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Stella Bergemann
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany
| | - Jessica Kowalski
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany
| | - Michael Huck
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany
| | - Christoph Bode
- Heart Center, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
24
|
Likhite N, Yadav V, Milliman EJ, Sopariwala DH, Lorca S, Narayana NP, Sheth M, Reineke EL, Giguère V, Narkar V. Loss of Estrogen-Related Receptor Alpha Facilitates Angiogenesis in Endothelial Cells. Mol Cell Biol 2019; 39:e00411-18. [PMID: 30602497 PMCID: PMC6379583 DOI: 10.1128/mcb.00411-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/04/2018] [Accepted: 12/11/2018] [Indexed: 01/08/2023] Open
Abstract
Estrogen-related receptors (ERRs) have emerged as major metabolic regulators in various tissues. However, their expression and function in the vasculature remains unknown. Here, we report the transcriptional program and cellular function of ERRα in endothelial cells (ECs), a cell type with a multifaceted role in vasculature. Of the three ERR subtypes, ECs exclusively express ERRα. Gene expression profiling of ECs lacking ERRα revealed that ERRα predominantly acts as a transcriptional repressor, targeting genes linked with angiogenesis, cell migration, and cell adhesion. ERRα-deficient ECs exhibit decreased proliferation but increased migration and tube formation. ERRα depletion increased basal as well as vascular endothelial growth factor A (VEGFA)- and ANG1/2-stimulated angiogenic sprouting in endothelial spheroids. Moreover, retinal angiogenesis is enhanced in ERRα knockout mice compared to that in wild-type mice. Surprisingly, ERRα is dispensable for the regulation of its classic targets, such as metabolism, mitochondrial biogenesis, and cellular respiration in the ECs. ERRα is enriched at the promoters of angiogenic, migratory, and cell adhesion genes. Further, VEGFA increased ERRα recruitment to angiogenesis-associated genes and simultaneously decreased their expression. Despite increasing its gene occupancy, proangiogenic stimuli decrease ERRα expression in ECs. Our work shows that endothelial ERRα plays a repressive role in angiogenesis and potentially fine-tunes growth factor-mediated angiogenesis.
Collapse
Affiliation(s)
- Neah Likhite
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Vikas Yadav
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | | | - Danesh H Sopariwala
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Sabina Lorca
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Nithya P Narayana
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Megha Sheth
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
- Department of Biochemistry and Cell Biology, Rice University, Houston, Texas, USA
| | - Erin L Reineke
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Vincent Giguère
- Department of Biochemistry, Medicine and Oncology, Faculty of Medicine, Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Vihang Narkar
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
- Integrative Biology and Pharmacology, The University of Texas McGovern Medical School, Houston, Texas, USA
- Graduate School of Biomedical Sciences at The University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
25
|
Lother A, Deng L, Huck M, Fürst D, Kowalski J, Esser JS, Moser M, Bode C, Hein L. Endothelial cell mineralocorticoid receptors oppose VEGF-induced gene expression and angiogenesis. J Endocrinol 2019; 240:15-26. [PMID: 30400069 DOI: 10.1530/joe-18-0494] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 09/24/2018] [Indexed: 12/29/2022]
Abstract
Aldosterone is a key factor in adverse cardiovascular remodeling by acting on the mineralocorticoid receptor (MR) in different cell types. Endothelial MR activation mediates hypertrophy, inflammation and fibrosis. Cardiovascular remodeling is often accompanied by impaired angiogenesis, which is a risk factor for the development of heart failure. In this study, we evaluated the impact of MR in endothelial cells on angiogenesis. Deoxycorticosterone acetate (DOCA)-induced hypertension was associated with capillary rarefaction in the heart of WT mice but not of mice with cell type-specific MR deletion in endothelial cells. Consistently, endothelial MR deletion prevented the inhibitory effect of aldosterone on the capillarization of subcutaneously implanted silicon tubes and on capillary sprouting from aortic ring segments. We examined MR-dependent gene expression in cultured endothelial cells by RNA-seq and identified a cluster of differentially regulated genes related to angiogenesis. We found opposing effects on gene expression when comparing activation of the mineralocorticoid receptor in ECs to treatment with vascular endothelial growth factor (VEGF), a potent activator of angiogenesis. In conclusion, we demonstrate here that activation of endothelial cell MR impaired angiogenic capacity and lead to capillary rarefaction in a mouse model of MR-driven hypertension. MR activation opposed VEGF-induced gene expression leading to the dysregulation of angiogenesis-related gene networks in endothelial cells. Our findings underscore the pivotal role of endothelial cell MR in the pathophysiology of hypertension and related heart disease.
Collapse
Affiliation(s)
- Achim Lother
- A Lother, Institute of experimental and clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Lisa Deng
- L Deng, Institute of experimental and clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Michael Huck
- M Huck, Institute of experimental and clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - David Fürst
- D Fürst, Institute of experimental and clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Jessica Kowalski
- J Kowalski, Institute of experimental and clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Jennifer Susanne Esser
- J Esser, Heart Center, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Martin Moser
- M Moser, Heart Center, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- C Bode, Heart Center, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Lutz Hein
- L Hein, Institute of experimental and clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
26
|
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
|
27
|
Lipshultz SE, Herman EH. Anthracycline cardiotoxicity: the importance of horizontally integrating pre-clinical and clinical research. Cardiovasc Res 2018; 114:205-209. [PMID: 29272330 PMCID: PMC5852510 DOI: 10.1093/cvr/cvx246] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Steven E Lipshultz
- Department of Pediatrics, Wayne State University School of Medicine, Karmanos Cancer Institute, and Children’s Hospital of Michigan, 3901 Beaubien Boulevard, Pediatric Administration-T121A, Detroit, MI 48201, USA
| | - Eugene H Herman
- Toxicology and Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, The National Cancer Institute, Rockville, MD 20850-9734, USA
| |
Collapse
|
28
|
Tamargo M, Tamargo J. Future drug discovery in renin-angiotensin-aldosterone system intervention. Expert Opin Drug Discov 2017; 12:827-848. [PMID: 28541811 DOI: 10.1080/17460441.2017.1335301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Renin-angiotensin-aldosterone system inhibitors (RAASIs), including angiotensin-converting enzyme inhibitors, angiotensin AT1 receptor blockers and mineralocorticoid receptor antagonists (MRAs), are the cornerstone for the treatment of cardiovascular and renal diseases. Areas covered: The authors searched MEDLINE, PubMed and ClinicalTrials.gov to identify eligible full-text English language papers. Herein, the authors discuss AT2-receptor agonists and ACE2/angiotensin-(1-7)/Mas-receptor axis modulators, direct renin inhibitors, brain aminopeptidase A inhibitors, biased AT1R blockers, chymase inhibitors, multitargeted drugs, vaccines and aldosterone receptor antagonists as well as aldosterone synthase inhibitors. Expert opinion: Preclinical studies have demonstrated that activation of the protective axis of the RAAS represents a novel therapeutic strategy for treating cardiovascular and renal diseases, but there are no clinical trials supporting our expectations. Non-steroidal MRAs might become the third-generation of MRAs for the treatment of heart failure, diabetes mellitus and chronic kidney disease. The main challenge for these new drugs is that conventional RAASIs are safe, effective and cheap generics. Thus, the future of new RAASIs will be directed by economical/strategic reasons.
Collapse
Affiliation(s)
- Maria Tamargo
- a Department of Cardiology , Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV , Madrid , Spain
| | - Juan Tamargo
- b Department of Pharmacology , School of Medicine, University Complutense, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV , Madrid , Spain
| |
Collapse
|
29
|
Zhang YY, Li C, Yao GF, Du LJ, Liu Y, Zheng XJ, Yan S, Sun JY, Liu Y, Liu MZ, Zhang X, Wei G, Tong W, Chen X, Wu Y, Sun S, Liu S, Ding Q, Yu Y, Yin H, Duan SZ. Deletion of Macrophage Mineralocorticoid Receptor Protects Hepatic Steatosis and Insulin Resistance Through ERα/HGF/Met Pathway. Diabetes 2017; 66:1535-1547. [PMID: 28325853 PMCID: PMC5860190 DOI: 10.2337/db16-1354] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 03/13/2017] [Indexed: 12/20/2022]
Abstract
Although the importance of macrophages in nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM) has been recognized, how macrophages affect hepatocytes remains elusive. Mineralocorticoid receptor (MR) has been implicated to play important roles in NAFLD and T2DM. However, cellular and molecular mechanisms are largely unknown. We report that myeloid MR knockout (MRKO) improves glucose intolerance, insulin resistance, and hepatic steatosis in obese mice. Estrogen signaling is sufficient and necessary for such improvements. Hepatic gene and protein expression suggests that MRKO reduces hepatic lipogenesis and lipid storage. In the presence of estrogen, MRKO in macrophages decreases lipid accumulation and increases insulin sensitivity of hepatocytes through hepatocyte growth factor (HGF)/Met signaling. MR directly regulates estrogen receptor 1 (Esr1 [encoding ERα]) in macrophages. Knockdown of hepatic Met eliminates the beneficial effects of MRKO in female obese mice. These findings identify a novel MR/ERα/HGF/Met pathway that conveys metabolic signaling from macrophages to hepatocytes in hepatic steatosis and insulin resistance and provide potential new therapeutic strategies for NAFLD and T2DM.
Collapse
Affiliation(s)
- Yu-Yao Zhang
- Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Chao Li
- Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Gao-Feng Yao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Lin-Juan Du
- Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Yuan Liu
- Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Xiao-Jun Zheng
- Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Shuai Yan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Jian-Yong Sun
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Yan Liu
- Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Zhu Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Xiaoran Zhang
- Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Gang Wei
- Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenxin Tong
- Department of Infectious Diseases, Ren-Min Hospital of Wuhan University, Wuhan, China
| | - Xiaobei Chen
- Department of Infectious Diseases, Ren-Min Hospital of Wuhan University, Wuhan, China
| | - Yong Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA
| | - Shuyang Sun
- Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Suling Liu
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University, Shanghai, China
| | - Qiurong Ding
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Ying Yu
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Huiyong Yin
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Sun XN, Li C, Liu Y, Du LJ, Zeng MR, Zheng XJ, Zhang WC, Liu Y, Zhu M, Kong D, Zhou L, Lu L, Shen ZX, Yi Y, Du L, Qin M, Liu X, Hua Z, Sun S, Yin H, Zhou B, Yu Y, Zhang Z, Duan SZ. T-Cell Mineralocorticoid Receptor Controls Blood Pressure by Regulating Interferon-Gamma. Circ Res 2017; 120:1584-1597. [DOI: 10.1161/circresaha.116.310480] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/07/2017] [Accepted: 03/13/2017] [Indexed: 11/16/2022]
Abstract
Rationale:
Hypertension remains to be a global public health burden and demands novel intervention strategies such as targeting T cells and T-cell–derived cytokines. Mineralocorticoid receptor (MR) antagonists have been clinically used to treat hypertension. However, the function of T-cell MR in blood pressure (BP) regulation has not been elucidated.
Objective:
We aim to determine the role of T-cell MR in BP regulation and to explore the mechanism.
Methods and Results:
Using T-cell MR knockout mouse in combination with angiotensin II–induced hypertensive mouse model, we demonstrated that MR deficiency in T cells strikingly decreased both systolic and diastolic BP and attenuated renal and vascular damage. Flow cytometric analysis showed that T-cell MR knockout mitigated angiotensin II–induced accumulation of interferon-gamma (IFN-γ)–producing T cells, particularly CD8
+
population, in both kidneys and aortas. Similarly, eplerenone attenuated angiotensin II–induced elevation of BP and accumulation of IFN-γ–producing T cells in wild-type mice. In cultured CD8
+
T cells, T-cell MR knockout suppressed IFN-γ expression whereas T-cell MR overexpression and aldosterone both enhanced IFN-γ expression. At the molecular level, MR interacted with NFAT1 (nuclear factor of activated T-cells 1) and activator protein-1 in T cells. Finally, T-cell MR overexpressing mice manifested more elevated BP compared with control mice after angiotensin II infusion and such difference was abolished by IFN-γ–neutralizing antibodies.
Conclusions:
MR may interact with NFAT1 and activator protein-1 to control IFN-γ in T cells and to regulate target organ damage and ultimately BP. Targeting MR in T cells specifically may be an effective novel approach for hypertension treatment.
Collapse
Affiliation(s)
- Xue-Nan Sun
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Chao Li
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Yuan Liu
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Lin-Juan Du
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Meng-Ru Zeng
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Xiao-Jun Zheng
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Wu-Chang Zhang
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Yan Liu
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Mingjiang Zhu
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Deping Kong
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Li Zhou
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Limin Lu
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Zhu-Xia Shen
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Yi Yi
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Lili Du
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Mu Qin
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Xu Liu
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Zichun Hua
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Shuyang Sun
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Huiyong Yin
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Bin Zhou
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Ying Yu
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Zhiyuan Zhang
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| | - Sheng-Zhong Duan
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Ninth People’s Hospital, School of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.-Z.D.), Shanghai Key Laboratory of Stomatology (X.-N.S., C.L., Y.L., L.-J.D., M.-R.Z., X.-J.Z., W.-C.Z., Y.L., S.S., Z.Z., S.-Z.D.), Department of Cardiology, Shanghai Chest Hospital (Y.Y., L.D., M.Q., X.L.), and Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital (S.S., Z.Z
| |
Collapse
|
31
|
Chadwick JA, Hauck JS, Gomez-Sanchez CE, Gomez-Sanchez EP, Rafael-Fortney JA. Gene expression effects of glucocorticoid and mineralocorticoid receptor agonists and antagonists on normal human skeletal muscle. Physiol Genomics 2017; 49:277-286. [PMID: 28432191 DOI: 10.1152/physiolgenomics.00128.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/10/2017] [Accepted: 04/17/2017] [Indexed: 12/28/2022] Open
Abstract
Mineralocorticoid and glucocorticoid receptors are closely related steroid hormone receptors that regulate gene expression through many of the same hormone response elements. However, their transcriptional activities and effects in skeletal muscles are largely unknown. We recently identified mineralocorticoid receptors (MR) in skeletal muscles after finding that combined treatment with the angiotensin-converting enzyme inhibitor lisinopril and MR antagonist spironolactone was therapeutic in Duchenne muscular dystrophy mouse models. The glucocorticoid receptor (GR) agonist prednisolone is the current standard-of-care treatment for Duchenne muscular dystrophy because it prolongs ambulation, likely due to its anti-inflammatory effects. However, data on whether glucocorticoids have a beneficial or detrimental direct effect on skeletal muscle are controversial. Here, we begin to define the gene expression profiles in normal differentiated human skeletal muscle myotubes treated with MR and GR agonists and antagonists. The MR agonist aldosterone and GR agonist prednisolone had highly overlapping gene expression profiles, supporting the notion that prednisolone acts as both a GR and MR agonist that may have detrimental effects on skeletal muscles. Co-incubations with aldosterone plus either nonspecific or selective MR antagonists, spironolactone or eplerenone, resulted in similar numbers of gene expression changes, suggesting that both drugs can block MR activation to a similar extent. Eplerenone treatment alone decreased a number of important muscle-specific genes. This information may be used to develop biomarkers to monitor clinical efficacy of MR antagonists or GR agonists in muscular dystrophy, develop a temporally coordinated treatment with both drugs, or identify novel therapeutics with more specific downstream targets.
Collapse
Affiliation(s)
- Jessica A Chadwick
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio
| | - J Spencer Hauck
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Celso E Gomez-Sanchez
- Department of Internal Medicine, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - Elise P Gomez-Sanchez
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jill A Rafael-Fortney
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio;
| |
Collapse
|
32
|
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]
|
33
|
Kolkhof P, Jaisser F, Kim SY, Filippatos G, Nowack C, Pitt B. Steroidal and Novel Non-steroidal Mineralocorticoid Receptor Antagonists in Heart Failure and Cardiorenal Diseases: Comparison at Bench and Bedside. Handb Exp Pharmacol 2017; 243:271-305. [PMID: 27830348 DOI: 10.1007/164_2016_76] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Characterization of mice with cell-specific deletion or overexpression of the mineralocorticoid receptor (MR) shed a new light on its role in health and disease. Pathophysiological MR activation contributes to a plethora of deleterious molecular mechanisms in the development of cardiorenal diseases like chronic kidney disease (CKD) and heart failure (HF). Accordingly, the available steroidal MR antagonists (MRAs) spironolactone (first generation MRA) and eplerenone (second generation MRA) have been shown to be effective in reducing cardiovascular (CV) mortality and morbidity in patients with chronic HF and a reduced left ventricular ejection fraction (HFrEF). However, they remain underutilized, in large part owing to the risk inducing severe adverse events including hyperkalemia and worsening of kidney function, particularly when given on top of inhibitors of the renin angiotensin system (RAS) to patients with concomitant kidney dysfunction. Novel, potent, and selective non-steroidal MRAs (third generation) were identified in drug discovery campaigns and a few entered clinical development recently. One of these is finerenone with different physicochemical, pharmacokinetics, and pharmacological properties in comparison with the steroidal MRAs. Available data from five clinical phase II trials with finerenone in more than 2,000 patients with HF and additional CKD and/or diabetes as well as in patients with diabetic kidney disease demonstrated that neither hyperkalemia nor reductions in kidney function were limiting factors to its use. Moreover, finerenone demonstrated a nominally improved outcome compared to eplerenone in a phase IIb trial with 1,066 patients with HFrEF and concomitant type 2 diabetes mellitus (T2DM) and/or CKD.
Collapse
Affiliation(s)
- Peter Kolkhof
- Drug Discovery, Cardiology Research, Bayer Pharma AG, Building 500, Aprather Weg 18a, 42096, Wuppertal, Germany.
| | - Frederic Jaisser
- INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, Paris, France
| | - So-Young Kim
- Clinical Development, Bayer Pharma AG, 42096, Wuppertal, Germany
| | - Gerasimos Filippatos
- Department of Cardiology, National and Kapodistrian University of Athens, School of Medicine, Attikon University Hospital, Rimini 1, Haidari, Athens, 12462, Greece
| | - Christina Nowack
- Clinical Development, Bayer Pharma AG, 42096, Wuppertal, Germany
| | - Bertram Pitt
- University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
34
|
Shen ZX, Chen XQ, Sun XN, Sun JY, Zhang WC, Zheng XJ, Zhang YY, Shi HJ, Zhang JW, Li C, Wang J, Liu X, Duan SZ. Mineralocorticoid Receptor Deficiency in Macrophages Inhibits Atherosclerosis by Affecting Foam Cell Formation and Efferocytosis. J Biol Chem 2016; 292:925-935. [PMID: 27881672 DOI: 10.1074/jbc.m116.739243] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 10/15/2016] [Indexed: 12/12/2022] Open
Abstract
Mineralocorticoid receptor (MR) has been considered as a potential target for treating atherosclerosis. However, the cellular and molecular mechanisms are not completely understood. We aim to explore the functions and mechanisms of macrophage MR in atherosclerosis. Atherosclerosis-susceptible LDLRKO chimeric mice with bone marrow cells from floxed control mice or from myeloid MR knock-out (MRKO) mice were generated and fed with high cholesterol diet. Oil red O staining showed that MRKO decreased atherosclerotic lesion area in LDLRKO mice. In another mouse model of atherosclerosis, MRKO/APOEKO mice and floxed control/APOEKO mice were generated and treated with angiotensin II. Similarly, MRKO inhibited the atherosclerotic lesion area in APOEKO mice. Histological analysis showed that MRKO increased collagen coverage and decreased necrosis and macrophage accumulation in the lesions. In vitro results demonstrated that MRKO suppressed macrophage foam cell formation and up-regulated the expression of genes involved in cholesterol efflux. Furthermore, MRKO decreased accumulation of apoptotic cells and increased effective efferocytosis in atherosclerotic lesions. In vitro study further revealed that MRKO increased the phagocytic index of macrophages without affecting their apoptosis. In conclusion, MRKO reduces high cholesterol- or angiotensin II-induced atherosclerosis and favorably changes plaque composition, likely improving plaque stability. Mechanistically, MR deficiency suppresses macrophage foam cell formation and up-regulates expression of genes related to cholesterol efflux, as well as increases effective efferocytosis and phagocytic capacity of macrophages.
Collapse
Affiliation(s)
- Zhu-Xia Shen
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,the Department of Cardiology, Jing'an District Centre Hospital of Shanghai, Huashan Hospital Jing'an Branch, Fudan University, Shanghai 200040, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031, China, and
| | - Xiao-Qing Chen
- the Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xue-Nan Sun
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031, China, and
| | - Jian-Yong Sun
- the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031, China, and
| | - Wu-Chang Zhang
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiao-Jun Zheng
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031, China, and
| | - Yu-Yao Zhang
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031, China, and
| | - Huan-Jing Shi
- the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031, China, and
| | - Jia-Wei Zhang
- the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031, China, and
| | - Chao Li
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031, China, and
| | - Jun Wang
- the Department of Cardiology, Jing'an District Centre Hospital of Shanghai, Huashan Hospital Jing'an Branch, Fudan University, Shanghai 200040, China
| | - Xu Liu
- the Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Sheng-Zhong Duan
- From the Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China,
| |
Collapse
|
35
|
Lother A, Hein L. Pharmacology of heart failure: From basic science to novel therapies. Pharmacol Ther 2016; 166:136-49. [PMID: 27456554 DOI: 10.1016/j.pharmthera.2016.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/08/2016] [Indexed: 01/10/2023]
Abstract
Chronic heart failure is one of the leading causes for hospitalization in the United States and Europe, and is accompanied by high mortality. Current pharmacological therapy of chronic heart failure with reduced ejection fraction is largely based on compounds that inhibit the detrimental action of the adrenergic and the renin-angiotensin-aldosterone systems on the heart. More than one decade after spironolactone, two novel therapeutic principles have been added to the very recently released guidelines on heart failure therapy: the HCN-channel inhibitor ivabradine and the combined angiotensin and neprilysin inhibitor valsartan/sacubitril. New compounds that are in phase II or III clinical evaluation include novel non-steroidal mineralocorticoid receptor antagonists, guanylate cyclase activators or myosine activators. A variety of novel candidate targets have been identified and the availability of gene transfer has just begun to accelerate translation from basic science to clinical application. This review provides an overview of current pharmacology and pharmacotherapy in chronic heart failure at three stages: the updated clinical guidelines of the American Heart Association and the European Society of Cardiology, new drugs which are in clinical development, and finally innovative drug targets and their mechanisms in heart failure which are emerging from preclinical studies will be discussed.
Collapse
Affiliation(s)
- Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Heart Center, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
36
|
Affiliation(s)
- Achim Lother
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, (A.L., L.H.), Heart Center, Department of Cardiology and Angiology I, (A.L.), and BIOSS Centre for Biological Signaling Studies (L.H.), University of Freiburg, Germany
| | - Lutz Hein
- From the Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, (A.L., L.H.), Heart Center, Department of Cardiology and Angiology I, (A.L.), and BIOSS Centre for Biological Signaling Studies (L.H.), University of Freiburg, Germany
| |
Collapse
|
37
|
Sun JY, Li C, Shen ZX, Zhang WC, Ai TJ, Du LJ, Zhang YY, Yao GF, Liu Y, Sun S, Naray-Fejes-Toth A, Fejes-Toth G, Peng Y, Chen M, Liu X, Tao J, Zhou B, Yu Y, Guo F, Du J, Duan SZ. Mineralocorticoid Receptor Deficiency in Macrophages Inhibits Neointimal Hyperplasia and Suppresses Macrophage Inflammation Through SGK1-AP1/NF-κB Pathways. Arterioscler Thromb Vasc Biol 2016; 36:874-85. [DOI: 10.1161/atvbaha.115.307031] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/19/2016] [Indexed: 01/08/2023]
Abstract
Objective—
Restenosis after percutaneous coronary intervention remains to be a serious medical problem. Although mineralocorticoid receptor (MR) has been implicated as a potential target for treating restenosis, the cellular and molecular mechanisms are largely unknown. This study aims to explore the functions of macrophage MR in neointimal hyperplasia and to delineate the molecular mechanisms.
Approach and Results—
Myeloid MR knockout (MMRKO) mice and controls were subjected to femoral artery injury. MMRKO reduced intima area and intima/media ratio, Ki67- and BrdU-positive vascular smooth muscle cells, expression of proinflammatory molecules, and macrophage accumulation in injured arteries. MMRKO macrophages migrated less in culture. MMRKO decreased Ki67- and BrdU-positive macrophages in injured arteries. MMRKO macrophages were less Ki67-positive in culture. Conditioned media from MMRKO macrophages induced less migration, Ki67 positivity, and proinflammatory gene expression of vascular smooth muscle cells. After lipopolysaccharide treatment, MMRKO macrophages had decreased p-cFos and p-cJun compared with control macrophages, suggesting suppressed activation of activator protein-1 (AP1). Nuclear factor-κB (NF-κB) pathway was also inhibited by MMRKO, manifested by decreased p-IκB kinase-β and p-IκBα, increased IκBα expression, decreased nuclear translocation of p65 and p50, as welll as decreased phosphorylation and expression of p65. Finally, overexpression of serum-and-glucocorticoid-inducible-kinase-1 (SGK1) attenuated the effects of MR deficiency in macrophages.
Conclusions—
Selective deletion of MR in myeloid cells limits macrophage accumulation and vascular inflammation and, therefore, inhibits neointimal hyperplasia and vascular remodeling. Mechanistically, MR deficiency suppresses migration and proliferation of macrophages and leads to less vascular smooth muscle cell activation. At the molecular level, MR deficiency suppresses macrophage inflammatory response via SGK1-AP1/NF-κB pathways.
Collapse
Affiliation(s)
- Jian-Yong Sun
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Chao Li
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Zhu-Xia Shen
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Wu-Chang Zhang
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Tang-Jun Ai
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Lin-Juan Du
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Yu-Yao Zhang
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Gao-Feng Yao
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Yan Liu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Shuyang Sun
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Aniko Naray-Fejes-Toth
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Geza Fejes-Toth
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Yong Peng
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Mao Chen
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Xiaojing Liu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Jun Tao
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Bin Zhou
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Ying Yu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Feifan Guo
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Jie Du
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| | - Sheng-Zhong Duan
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (J.-Y.S., C.L., Z.-X.S., W.-C.Z., T.-J.A., L.-J.D., Y.-Y.Z., G.-F.Y., Y.L., B.Z., Y.Y., F.G., S.-Z.D.); Shanghai Key Laboratory of Stomatology, Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
| |
Collapse
|
38
|
Lother A, Fürst D, Bergemann S, Gilsbach R, Grahammer F, Huber TB, Hilgendorf I, Bode C, Moser M, Hein L. Deoxycorticosterone Acetate/Salt–Induced Cardiac But Not Renal Injury Is Mediated By Endothelial Mineralocorticoid Receptors Independently From Blood Pressure. Hypertension 2016; 67:130-8. [DOI: 10.1161/hypertensionaha.115.06530] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 10/12/2015] [Indexed: 12/23/2022]
Affiliation(s)
- Achim Lother
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - David Fürst
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Stella Bergemann
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Ralf Gilsbach
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Florian Grahammer
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Tobias B. Huber
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Martin Moser
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Lutz Hein
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| |
Collapse
|
39
|
Gaudet HM, Cheng SB, Christensen EM, Filardo EJ. The G-protein coupled estrogen receptor, GPER: The inside and inside-out story. Mol Cell Endocrinol 2015; 418 Pt 3:207-19. [PMID: 26190834 DOI: 10.1016/j.mce.2015.07.016] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 07/15/2015] [Accepted: 07/15/2015] [Indexed: 02/06/2023]
Abstract
GPER possesses structural and functional characteristics shared by members of the G-protein-coupled receptor (GPCR) superfamily, the largest class of plasma membrane receptors. This newly appreciated estrogen receptor is localized predominately within intracellular membranes in most, but not all, cell types and its surface expression is modulated by steroid hormones and during tissue injury. An intracellular staining pattern is not unique among GPCRs, which employ a diverse array of molecular mechanisms that restrict cell surface expression and effectively regulating receptor binding and activation. The finding that GPER displays an intracellular predisposition has created some confusion as the estrogen-inducible transcription factors, ERα and ERβ, also reside intracellularly, and has led to complex suggestions of receptor interaction. GPER undergoes constitutive retrograde trafficking from the plasma membrane to the endoplasmic reticulum and recent studies indicate its interaction with PDZ binding proteins that sort transmembrane receptors to synaptosomes and endosomes. Genetic targeting and selective ligand approaches as well as cell models that express GPER in the absence of ERs clearly supports GPER as a bonafide "stand alone" receptor. Here, the molecular details that regulate GPER action, its cell biological activities and its implicated roles in physiological and pathological processes are reviewed.
Collapse
Affiliation(s)
- H M Gaudet
- Wheaton College, Department of Chemistry, Norton, MA, 02766, USA
| | - S B Cheng
- Women & Infants Hospital, Brown University, Providence, RI, 02903, USA
| | - E M Christensen
- Wheaton College, Department of Chemistry, Norton, MA, 02766, USA
| | - E J Filardo
- Rhode Island Hospital, Brown University, Providence, RI, 02903, USA.
| |
Collapse
|
40
|
Chadwick JA, Hauck JS, Lowe J, Shaw JJ, Guttridge DC, Gomez-Sanchez CE, Gomez-Sanchez EP, Rafael-Fortney JA. Mineralocorticoid receptors are present in skeletal muscle and represent a potential therapeutic target. FASEB J 2015; 29:4544-54. [PMID: 26178166 DOI: 10.1096/fj.15-276782] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 06/30/2015] [Indexed: 02/06/2023]
Abstract
Early treatment with heart failure drugs lisinopril and spironolactone improves skeletal muscle pathology in Duchenne muscular dystrophy (DMD) mouse models. The angiotensin converting enzyme inhibitor lisinopril and mineralocorticoid receptor (MR) antagonist spironolactone indirectly and directly target MR. The presence and function of MR in skeletal muscle have not been explored. MR mRNA and protein are present in all tested skeletal muscles from both wild-type mice and DMD mouse models. MR expression is cell autonomous in both undifferentiated myoblasts and differentiated myotubes from mouse and human skeletal muscle cultures. To test for MR function in skeletal muscle, global gene expression analysis was conducted on human myotubes treated with MR agonist (aldosterone; EC50 1.3 nM) or antagonist (spironolactone; IC50 1.6 nM), and 53 gene expression differences were identified. Five differences were conserved in quadriceps muscles from dystrophic mice treated with spironolactone plus lisinopril (IC50 0.1 nM) compared with untreated controls. Genes down-regulated more than 2-fold by MR antagonism included FOS, ANKRD1, and GADD45B, with known roles in skeletal muscle, in addition to NPR3 and SERPINA3, bona fide targets of MR in other tissues. MR is a novel drug target in skeletal muscle and use of clinically safe antagonists may be beneficial for muscle diseases.
Collapse
Affiliation(s)
- Jessica A Chadwick
- *Department of Molecular and Cellular Biochemistry, Department of Physiology and Cell Biology, Department of Molecular Virology, Immunology, and Medical Genetics College of Medicine, The Ohio State University, Columbus, Ohio, USA; and Department of Internal Medicine and Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - J Spencer Hauck
- *Department of Molecular and Cellular Biochemistry, Department of Physiology and Cell Biology, Department of Molecular Virology, Immunology, and Medical Genetics College of Medicine, The Ohio State University, Columbus, Ohio, USA; and Department of Internal Medicine and Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Jeovanna Lowe
- *Department of Molecular and Cellular Biochemistry, Department of Physiology and Cell Biology, Department of Molecular Virology, Immunology, and Medical Genetics College of Medicine, The Ohio State University, Columbus, Ohio, USA; and Department of Internal Medicine and Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Jeremiah J Shaw
- *Department of Molecular and Cellular Biochemistry, Department of Physiology and Cell Biology, Department of Molecular Virology, Immunology, and Medical Genetics College of Medicine, The Ohio State University, Columbus, Ohio, USA; and Department of Internal Medicine and Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Denis C Guttridge
- *Department of Molecular and Cellular Biochemistry, Department of Physiology and Cell Biology, Department of Molecular Virology, Immunology, and Medical Genetics College of Medicine, The Ohio State University, Columbus, Ohio, USA; and Department of Internal Medicine and Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Celso E Gomez-Sanchez
- *Department of Molecular and Cellular Biochemistry, Department of Physiology and Cell Biology, Department of Molecular Virology, Immunology, and Medical Genetics College of Medicine, The Ohio State University, Columbus, Ohio, USA; and Department of Internal Medicine and Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Elise P Gomez-Sanchez
- *Department of Molecular and Cellular Biochemistry, Department of Physiology and Cell Biology, Department of Molecular Virology, Immunology, and Medical Genetics College of Medicine, The Ohio State University, Columbus, Ohio, USA; and Department of Internal Medicine and Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Jill A Rafael-Fortney
- *Department of Molecular and Cellular Biochemistry, Department of Physiology and Cell Biology, Department of Molecular Virology, Immunology, and Medical Genetics College of Medicine, The Ohio State University, Columbus, Ohio, USA; and Department of Internal Medicine and Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|