1
|
Mazzieri A, Timio F, Patera F, Trepiccione F, Bonomini M, Reboldi G. Aldosterone Synthase Inhibitors for Cardiorenal Protection: Ready for Prime Time? Kidney Blood Press Res 2024; 49:1041-1056. [PMID: 39557029 DOI: 10.1159/000542621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Aldosterone is the principal mineralocorticoid hormone and the final effector of the renin-angiotensin-aldosterone system. This hormone is primarily synthesized by the CYP11B2 enzyme and produced by the adrenal zona glomerulosa. Through genomic and non-genomic effects, it plays an important role in cardiovascular and renal disease. To counteract aldosterone-mediated damage, steroidal mineralocorticoid receptor antagonists are recommended by international guidelines, but endocrine side effects often limit their use in a substantial proportion of patients. Conversely, nonsteroidal mineralocorticoid receptor antagonists, with an improved selectivity and safety profile, are gaining a prominent position among therapeutic pillars. However, blocking the mineralocorticoid receptors does not completely inhibit aldosterone effects because of escape mechanisms and non-genomic activity. Thus, inhibiting aldosterone synthesis could be a promising strategy to prevent aldosterone-mediated cardiorenal damage. The limited specificity for CYP11B2 and side effects due to off-target activity hampered the development of first-generation aldosterone synthase inhibitors (ASIs). SUMMARY The development of highly specific ASIs led to successful clinical trials in patients with resistant and uncontrolled hypertension. Additionally, a recent randomized clinical trial showed a significant benefit of ASIs in patients with chronic kidney disease and albuminuria. KEY MESSAGES The strength of the clinical evidence collected so far is still limited, and larger outcome-based clinical trials are needed to confirm the promising role of ASIs in cardiorenal damage.
Collapse
Affiliation(s)
- Alessio Mazzieri
- Diabetes Clinic, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesca Timio
- Division of Nephrology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesco Patera
- Division of Nephrology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesco Trepiccione
- Department of Medical Translational Sciences, University of Campania, Naples, Italy
- Biogem, Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
| | - Mario Bonomini
- Nephrology and Dialysis Unit, Department of Medicine, G. D'Annunzio University, Chieti, Italy
- SS. Annunziata Hospital, Chieti, Italy
| | - Gianpaolo Reboldi
- Division of Nephrology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
2
|
Burgermeister E. Mitogen-Activated Protein Kinase and Nuclear Hormone Receptor Crosstalk in Cancer Immunotherapy. Int J Mol Sci 2023; 24:13661. [PMID: 37686465 PMCID: PMC10488039 DOI: 10.3390/ijms241713661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
The three major MAP-kinase (MAPK) pathways, ERK1/2, p38 and JNK/SAPK, are upstream regulators of the nuclear "hormone" receptor superfamily (NHRSF), with a prime example given by the estrogen receptor in breast cancer. These ligand-activated transcription factors exert non-genomic and genomic functions, where they are either post-translationally modified by phosphorylation or directly interact with components of the MAPK pathways, events that govern their transcriptional activity towards target genes involved in cell differentiation, proliferation, metabolism and host immunity. This molecular crosstalk takes place not only in normal epithelial or tumor cells, but also in a plethora of immune cells from the adaptive and innate immune system in the tumor-stroma tissue microenvironment. Thus, the drugability of both the MAPK and the NHRSF pathways suggests potential for intervention therapies, especially for cancer immunotherapy. This review summarizes the existing literature covering the expression and function of NHRSF subclasses in human tumors, both solid and leukemias, and their effects in combination with current clinically approved therapeutics against immune checkpoint molecules (e.g., PD1).
Collapse
Affiliation(s)
- Elke Burgermeister
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
| |
Collapse
|
3
|
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: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
4
|
Johnston JG, Welch AK, Cain BD, Sayeski PP, Gumz ML, Wingo CS. Aldosterone: Renal Action and Physiological Effects. Compr Physiol 2023; 13:4409-4491. [PMID: 36994769 PMCID: PMC11472823 DOI: 10.1002/cphy.c190043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Aldosterone exerts profound effects on renal and cardiovascular physiology. In the kidney, aldosterone acts to preserve electrolyte and acid-base balance in response to changes in dietary sodium (Na+ ) or potassium (K+ ) intake. These physiological actions, principally through activation of mineralocorticoid receptors (MRs), have important effects particularly in patients with renal and cardiovascular disease as demonstrated by multiple clinical trials. Multiple factors, be they genetic, humoral, dietary, or otherwise, can play a role in influencing the rate of aldosterone synthesis and secretion from the adrenal cortex. Normally, aldosterone secretion and action respond to dietary Na+ intake. In the kidney, the distal nephron and collecting duct are the main targets of aldosterone and MR action, which stimulates Na+ absorption in part via the epithelial Na+ channel (ENaC), the principal channel responsible for the fine-tuning of Na+ balance. Our understanding of the regulatory factors that allow aldosterone, via multiple signaling pathways, to function properly clearly implicates this hormone as central to many pathophysiological effects that become dysfunctional in disease states. Numerous pathologies that affect blood pressure (BP), electrolyte balance, and overall cardiovascular health are due to abnormal secretion of aldosterone, mutations in MR, ENaC, or effectors and modulators of their action. Study of the mechanisms of these pathologies has allowed researchers and clinicians to create novel dietary and pharmacological targets to improve human health. This article covers the regulation of aldosterone synthesis and secretion, receptors, effector molecules, and signaling pathways that modulate its action in the kidney. We also consider the role of aldosterone in disease and the benefit of mineralocorticoid antagonists. © 2023 American Physiological Society. Compr Physiol 13:4409-4491, 2023.
Collapse
Affiliation(s)
- Jermaine G Johnston
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Amanda K Welch
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Brian D Cain
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Peter P Sayeski
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - Michelle L Gumz
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Charles S Wingo
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| |
Collapse
|
5
|
Karst H, den Boon FS, Vervoort N, Adrian M, Kapitein LC, Joëls M. Non-genomic steroid signaling through the mineralocorticoid receptor: Involvement of a membrane-associated receptor? Mol Cell Endocrinol 2022; 541:111501. [PMID: 34740745 DOI: 10.1016/j.mce.2021.111501] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/05/2021] [Accepted: 10/27/2021] [Indexed: 12/25/2022]
Abstract
Corticosteroid receptors in the mammalian brain mediate genomic as well as non-genomic actions. Although receptors mediating genomic actions were already cloned 35 years ago, it remains unclear whether the same molecules are responsible for the non-genomic actions or that the latter involve a separate class of receptors. Here we focus on one type of corticosteroid receptors, i.e. the mineralocorticoid receptor (MR). We summarize some of the known properties and the current insight in the localization of the MR in peripheral cells and neurons, especially in relation to non-genomic signaling. Previous studies from our own and other labs provided evidence that MRs mediating non-genomic actions are identical to the ones involved in genomic signaling, but may be translocated to the plasma cell membrane instead of the nucleus. With fixed cell imaging and live cell imaging techniques we tried to visualize these presumed membrane-associated MRs, using antibodies or overexpression of MR-GFP in COS7 and hippocampal cultured neurons. Despite the physiological evidence for MR location in or close to the cell membrane, we could not convincingly visualize membrane localization of endogenous MRs or GFP-MR molecules. However, we did find punctae of labeled antibodies intracellularly, which might indicate transactivating spots of MR near the membrane. We also found some evidence for trafficking of MR via beta-arrestins. In beta-arrestin knockout mice, we didn't observe metaplasticity in the basolateral amygdala anymore, indicating that internalization of MRs could play a role during corticosterone activation. Furthermore, we speculate that membrane-associated MRs could act indirectly via activating other membrane located structures like e.g. GPER and/or receptor tyrosine kinases.
Collapse
Affiliation(s)
- Henk Karst
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands.
| | - Femke S den Boon
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Niek Vervoort
- University Utrecht, Faculty of Science, Division of Cell Biology, Utrecht, the Netherlands
| | - Max Adrian
- University Utrecht, Faculty of Science, Division of Cell Biology, Utrecht, the Netherlands
| | - Lukas C Kapitein
- University Utrecht, Faculty of Science, Division of Cell Biology, Utrecht, the Netherlands
| | - Marian Joëls
- Dept Translational Neuroscience, University Medical Center Utrecht, Utrecht University, the Netherlands; University Medical Center Groningen, University of Groningen, the Netherlands
| |
Collapse
|
6
|
Abstract
Steroid hormones bind receptors in the cell nucleus and in the cell membrane. The most widely studied class of steroid hormone receptors are the nuclear receptors, named for their function as ligand-dependent transcription factors in the cell nucleus. Nuclear receptors, such as estrogen receptor alpha, can also be anchored to the plasma membrane, where they respond to steroids by activating signaling pathways independent of their function as transcription factors. Steroids can also bind integral membrane proteins, such as the G protein-coupled estrogen receptor. Membrane estrogen and progestin receptors have been cloned and characterized in vitro and influence the development and function of many organ systems. Membrane androgen receptors were cloned and characterized in vitro, but their function as androgen receptors in vivo is unresolved. We review the identity and function of membrane proteins that bind estrogens, progestins, and androgens. We discuss evidence that membrane glucocorticoid and mineralocorticoid receptors exist, and whether glucocorticoid and mineralocorticoid nuclear receptors act at the cell membrane. In many cases, integral membrane steroid receptors act independently of nuclear steroid receptors, even though they may share a ligand.
Collapse
Affiliation(s)
- Lindsey S Treviño
- Department of Population Sciences, Division of Health Equities, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Daniel A Gorelick
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: Daniel A Gorelick, PhD, One Baylor Plaza, Alkek Building N1317.07, Houston, TX, 77030-3411, USA.
| |
Collapse
|
7
|
OPALS: A New Osimertinib Adjunctive Treatment of Lung Adenocarcinoma or Glioblastoma Using Five Repurposed Drugs. Cells 2021; 10:cells10051148. [PMID: 34068720 PMCID: PMC8151869 DOI: 10.3390/cells10051148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Pharmacological targeting aberrant activation of epidermal growth factor receptor tyrosine kinase signaling is an established approach to treating lung adenocarcinoma. Osimertinib is a tyrosine kinase approved and effective in treating lung adenocarcinomas that have one of several common activating mutations in epidermal growth factor receptor. The emergence of resistance to osimertinib after a year or two is the rule. We developed a five-drug adjuvant regimen designed to increase osimertinib’s growth inhibition and thereby delay the development of resistance. Areas of Uncertainty: Although the assembled preclinical data is strong, preclinical data and the following clinical trial results can be discrepant. The safety of OPALS drugs when used individually is excellent. We have no data from humans on their tolerability when used as an ensemble. That there is no data from the individual drugs to suspect problematic interaction does not exclude the possibility. Data Sources: All relevant PubMed.org articles on the OPALS drugs and corresponding pathophysiology of lung adenocarcinoma and glioblastoma were reviewed. Therapeutic Opinion: The five drugs of OPALS are in wide use in general medicine for non-oncology indications. OPALS uses the anti-protozoal drug pyrimethamine, the antihistamine cyproheptadine, the antibiotic azithromycin, the antihistamine loratadine, and the potassium sparing diuretic spironolactone. We show how these inexpensive and generically available drugs intersect with and inhibit lung adenocarcinoma growth drive. We also review data showing that both OPALS adjuvant drugs and osimertinib have data showing they may be active in suppressing glioblastoma growth.
Collapse
|
8
|
Ennis IL, Pérez NG. Cardiac Mineralocorticoid Receptor and the Na +/H + Exchanger: Spilling the Beans. Front Cardiovasc Med 2021; 7:614279. [PMID: 33553262 PMCID: PMC7854694 DOI: 10.3389/fcvm.2020.614279] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/01/2020] [Indexed: 01/08/2023] Open
Abstract
Current evidence reveals that cardiac mineralocorticoid receptor (MR) activation following myocardial stretch plays an important physiological role in adapting developed force to sudden changes in hemodynamic conditions. Its underlying mechanism involves a previously unknown nongenomic effect of the MR that triggers redox-mediated Na+/H+ exchanger (NHE1) activation, intracellular Na+ accumulation, and a consequent increase in Ca2+ transient amplitude through reverse Na+/Ca2+ exchange. However, clinical evidence assigns a detrimental role to MR activation in the pathogenesis of severe cardiac diseases such as congestive heart failure. This mini review is meant to present and briefly discuss some recent discoveries about locally triggered cardiac MR signals with the objective of shedding some light on its physiological but potentially pathological consequences in the heart.
Collapse
Affiliation(s)
- Irene Lucía Ennis
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas de la Plata, Universidad Nacional de La Plata, La Plata, Argentina
| | - Néstor Gustavo Pérez
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas de la Plata, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
9
|
Zi Shen Huo Luo Formula Prevents Aldosterone-Induced Cardiomyocyte Hypertrophy and Cardiac Fibroblast Proliferation by Regulating the Striatin-Mediated MR/EGFR/ERK Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9028047. [PMID: 33014117 PMCID: PMC7519188 DOI: 10.1155/2020/9028047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 08/01/2020] [Accepted: 09/01/2020] [Indexed: 11/22/2022]
Abstract
Inappropriate activation of the renin-angiotensin-aldosterone system (RAAS) is an important factor in the development of hypertension. Excessive aldosterone can lead to myocardial extracellular matrix collagen proliferation, fibrosis, and cardiomyocyte hypertrophy and aggravate maladaptive remodeling. The results of our previous clinical and animal experiments suggested that Zi Shen Huo Luo Formula (ZSHLF) combined with perindopril can effectively control the process of left ventricular hypertrophy (LVH). The purpose of this study was to investigate whether ZSHLF-treated serum inhibits the membrane localization of the striatin-mediated mineralocorticoid receptor (MR) and affects MR-mediated nongenomic effects and the downstream epidermal growth factor receptor (EGFR)/extracellular regulated kinase (ERK) signaling pathways, thereby improving aldosterone-induced myocardial remodeling. Serum containing ZSHLF was prepared and used to treat rat cardiomyocytes and cardiac fibroblasts in vitro after aldosterone induction and striatin knockdown by small interfering RNA (siRNA). Cell-based assays were carried out to determine the cardiomyocyte surface area and assess the proliferation rate and hydroxyproline secretion of cardiac fibroblasts. Quantitative real-time PCR (qRT-PCR), immunoprecipitation (IP), and Western blotting were performed to evaluate the striatin-mediated MR/EGFR/ERK signaling pathway. In the present study, ZSHLF attenuated the aldosterone-induced hypertrophy of cardiomyocytes and inhibited the proliferation and collagen synthesis of cardiac fibroblasts. ZSHLF also reduced striatin mRNA expression and inhibited striatin and MR binding, membrane MR protein expression, and EGFR and ERK1/2 phosphorylation. Furthermore, after striatin silencing with siRNA, some of the effects of ZSHLF were not changed significantly. In conclusion, ZSHLF inhibits the downstream EGFR/ERK signaling pathway by blocking the striatin-mediated membrane localization of MR, which may be an important molecular mechanism by which ZSHLF improves aldosterone-induced myocardial remodeling.
Collapse
|
10
|
Shen Y, Zhang J, Xie J, Liu J. In vitro assessment of corticosteroid effects of eight chiral herbicides. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART. B, PESTICIDES, FOOD CONTAMINANTS, AND AGRICULTURAL WASTES 2019; 55:91-102. [PMID: 31524045 DOI: 10.1080/03601234.2019.1665408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Information regarding the enantioselective endocrine disruption of chiral herbicides is scarce. This study assessed the disrupting effects of eight typical chiral herbicides on corticosteroids (including glucocorticoids and mineralocorticoids). Enantioselectivity of eight chiral herbicides were evaluated for their agonistic/antagonistic effects on glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) with CHOK1 cell line using reporter gene assay. Their influence on the production of corticosteroids were further investigated in H295R cell line using enzyme-linked immunosorbent assay (ELISA). None of the racemates or enantiomers of eight chiral herbicides exhibited GR or MR agonistic activity at non-cytotoxic concentrations. However, rac-propisochlor and S-imazamox antagonized cortisol-induced transactivation of GR by 21.79% and 38.73% at the concentration of 1.0 × 10-7 M and 1.0 × 10-6 M, respectively, and R-napropamide remarkably attenuated aldosterone-induced MR transactivation by 68.78% at 1.0 × 10-6 M. The secretion of cortisol was significantly restrained after treated with 1.0 × 10-6 M rac-propisochlor and rac-/R-napropamide at the concentration of 1.0 × 10-6 M by 26.49%, 30.10% and 35.27%, respectively, while this glucocorticoid was remarkably induced by 1.0 × 10-5 M rac-diclofop-methyl and its two enantiomers at the concentration of 1.0 × 10-5 M by 75.60%, 100.1% and 68.78%, respectively. Exposure to rac-propisochlor (1.0 × 10-6 M), S-diclofop-methyl (1.0 × 10-5 M) or rac-/S-/R- acetochlor (1.0 × 10-6 M) and rac-/S-/R-lactofen (1.0 × 10-6 M) inhibited the secretion of aldosterone by approximately 40%. Our findings suggested that chiral herbicides disrupted corticosteroid homeostasis in an enantioselective way. Therefore, more comprehensive screening is required to better understand the ecological and health risks of chiral pesticides.
Collapse
Affiliation(s)
- Yuqing Shen
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, China
| | - Jianyun Zhang
- Department of Public Health, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Jingqian Xie
- College of Marine Ecology and Environment, Shanghai Ocean University, Shanghai, China
| | - Jing Liu
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Mihailidou AS, Tzakos AG, Ashton AW. Non-Genomic Effects of Aldosterone. VITAMINS AND HORMONES 2019; 109:133-149. [DOI: 10.1016/bs.vh.2018.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
12
|
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:ijms19123764. [PMID: 30486399 PMCID: PMC6320977 DOI: 10.3390/ijms19123764] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [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
|
13
|
Ruhs S, Nolze A, Hübschmann R, Grossmann C. 30 YEARS OF THE MINERALOCORTICOID RECEPTOR: Nongenomic effects via the mineralocorticoid receptor. J Endocrinol 2017; 234:T107-T124. [PMID: 28348113 DOI: 10.1530/joe-16-0659] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 12/12/2022]
Abstract
The mineralocorticoid receptor (MR) belongs to the steroid hormone receptor family and classically functions as a ligand-dependent transcription factor. It is involved in water-electrolyte homeostasis and blood pressure regulation but independent from these effects also furthers inflammation, fibrosis, hypertrophy and remodeling in cardiovascular tissues. Next to genomic effects, aldosterone elicits very rapid actions within minutes that do not require transcription or translation and that occur not only in classical MR epithelial target organs like kidney and colon but also in nonepithelial tissues like heart, vasculature and adipose tissue. Most of these effects can be mediated by classical MR and its crosstalk with different signaling cascades. Near the plasma membrane, the MR seems to be associated with caveolin and striatin as well as with receptor tyrosine kinases like EGFR, PDGFR and IGF1R and G protein-coupled receptors like AT1 and GPER1, which then mediate nongenomic aldosterone effects. GPER1 has also been named a putative novel MR. There is a close interaction and functional synergism between the genomic and the nongenomic signaling so that nongenomic signaling can lead to long-term effects and support genomic actions. Therefore, understanding nongenomic aldosterone/MR effects is of potential relevance for modulating genomic aldosterone effects and may provide additional targets for intervention.
Collapse
Affiliation(s)
- Stefanie Ruhs
- Julius Bernstein Institute of PhysiologyMartin Luther University Halle-Wittenberg, Halle, Germany
| | - Alexander Nolze
- Julius Bernstein Institute of PhysiologyMartin Luther University Halle-Wittenberg, Halle, Germany
| | - Ralf Hübschmann
- Julius Bernstein Institute of PhysiologyMartin Luther University Halle-Wittenberg, Halle, Germany
| | - Claudia Grossmann
- Julius Bernstein Institute of PhysiologyMartin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
14
|
GPER is involved in the stimulatory effects of aldosterone in breast cancer cells and breast tumor-derived endothelial cells. Oncotarget 2016; 7:94-111. [PMID: 26646587 PMCID: PMC4807985 DOI: 10.18632/oncotarget.6475] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/22/2015] [Indexed: 12/16/2022] Open
Abstract
Aldosterone induces relevant effects binding to the mineralcorticoid receptor (MR), which acts as a ligand-gated transcription factor. Alternate mechanisms can mediate the action of aldosterone such as the activation of epidermal growth factor receptor (EGFR), MAPK/ERK, transcription factors and ion channels. The G-protein estrogen receptor (GPER) has been involved in the stimulatory effects of estrogenic signalling in breast cancer. GPER has been also shown to contribute to certain responses to aldosterone, however the role played by GPER and the molecular mechanisms implicated remain to be fully understood. Here, we evaluated the involvement of GPER in the stimulatory action exerted by aldosterone in breast cancer cells and breast tumor derived endothelial cells (B-TEC). Competition assays, gene expression and silencing studies, immunoblotting and immunofluorescence experiments, cell proliferation and migration were performed in order to provide novel insights into the role of GPER in the aldosterone-activated signalling. Our results demonstrate that aldosterone triggers the EGFR/ERK transduction pathway in a MR- and GPER-dependent manner. Aldosterone does not bind to GPER, it however induces the direct interaction between MR and GPER as well as between GPER and EGFR. Next, we ascertain that the up-regulation of the Na+/H+ exchanger-1 (NHE-1) induced by aldosterone involves MR and GPER. Biologically, both MR and GPER contribute to the proliferation and migration of breast and endothelial cancer cells mediated by NHE-1 upon aldosterone exposure. Our data further extend the current knowledge on the molecular mechanisms through which GPER may contribute to the stimulatory action elicited by aldosterone in breast cancer.
Collapse
|
15
|
Barton M, Meyer MR. Nicolaus Copernicus and the rapid vascular responses to aldosterone. Trends Endocrinol Metab 2015; 26:396-8. [PMID: 26088671 DOI: 10.1016/j.tem.2015.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/12/2015] [Accepted: 05/13/2015] [Indexed: 12/31/2022]
Abstract
For decades, rapid steroid responses initiated by membrane receptors have been a primary research focus. G protein-coupled estrogen receptor (GPER) is activated by 17β-estradiol and participates in functional crosstalk with other steroid receptors. With reference to the physician and astronomer Nicolaus Copernicus (1473-1543), who used rigorous scientific approaches to shift paradigms and change dogma, we discuss whether GPER can also be considered an aldosterone receptor.
Collapse
Affiliation(s)
- Matthias Barton
- Molecular Internal Medicine, University of Zurich, Switzerland.
| | | |
Collapse
|
16
|
Ashton AW, Le TYL, Gomez-Sanchez CE, Morel-Kopp MC, McWhinney B, Hudson A, Mihailidou AS. Role of Nongenomic Signaling Pathways Activated by Aldosterone During Cardiac Reperfusion Injury. Mol Endocrinol 2015; 29:1144-55. [PMID: 26121234 DOI: 10.1210/me.2014-1410] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aldosterone (Aldo) activates both genomic and nongenomic signaling pathways in the cardiovascular system. Activation of genomic signaling pathways contributes to the adverse cardiac actions of Aldo during reperfusion injury; however, the extent nongenomic signaling pathways contribute has been difficult to identify due to lack of a specific ligand that activates only nongenomic signaling pathways. Using a pegylated aldosterone analog, aldosterone-3-carboxymethoxylamine-TFP ester conjugated to methoxypegylated amine (Aldo-PEG), we are able for the first time to distinguish between nongenomic and genomic cardiac actions of Aldo. We confirm Aldo-PEG activates phosphorylation of ERK1/2 in rat cardiomyocyte H9c2 cells similar to Aldo and G protein-coupled receptor 30 (GPR30 or GPER) agonist G1. GPER antagonist, G36, but not mineralocorticoid receptor (MR) antagonist spironolactone, prevented ERK1/2 phosphorylation by Aldo, Aldo-PEG, and G1. The selective nongenomic actions of Aldo-PEG are confirmed, with Aldo-PEG increasing superoxide production in H9c2 cells to similar levels as Aldo but having no effect on subcellular localization of MR. Striatin serves as a scaffold for GPER and MR, with GPER antagonist G36, but not spironolactone, restoring MR-striatin complexes. Aldo-PEG had no effect on MR-dependent transcriptional activation, whereas Aldo increased transcript levels of serum-regulated kinase 1 and plasminogen activator inhibitor-1. Using our ex vivo experimental rat model of myocardial infarction, we found aggravated infarct size and apoptosis by Aldo but not Aldo-PEG. Our studies confirm that in the heart, activation of nongenomic signaling pathways alone are not sufficient to trigger the deleterious effects of aldosterone during myocardial reperfusion injury.
Collapse
Affiliation(s)
- Anthony W Ashton
- Cardiovascular and Hormonal Research Laboratory, Department of Cardiology (T.Y.L.L., A.S.M.), Division of Perinatal Research (A.W.A.), Northern Blood Research Centre and Department of Haematology and Transfusion Medicine (M.-C.M.-K.), Sydney Neuro-Oncology Group and Bill Walsh Translational Cancer Research Laboratory (A.H.), Royal North Shore Hospital and Kolling Institute (A.W.A., T.Y.L.L., M.-C.M.-K., A.H., A.S.M.), Royal North Shore Hospital and The University of Sydney, Sydney 2065, Australia; Division of Endocrinology, G. V. (Sonny) Montgomery Veterans Affairs Medical Center and University of Mississippi Medical Center (C.E.G.-S.), Jackson, Mississippi 39216; and Analytical Chemistry Unit (B.M.), Pathology Queensland, Health Services Support Agency, Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| | - Thi Y L Le
- Cardiovascular and Hormonal Research Laboratory, Department of Cardiology (T.Y.L.L., A.S.M.), Division of Perinatal Research (A.W.A.), Northern Blood Research Centre and Department of Haematology and Transfusion Medicine (M.-C.M.-K.), Sydney Neuro-Oncology Group and Bill Walsh Translational Cancer Research Laboratory (A.H.), Royal North Shore Hospital and Kolling Institute (A.W.A., T.Y.L.L., M.-C.M.-K., A.H., A.S.M.), Royal North Shore Hospital and The University of Sydney, Sydney 2065, Australia; Division of Endocrinology, G. V. (Sonny) Montgomery Veterans Affairs Medical Center and University of Mississippi Medical Center (C.E.G.-S.), Jackson, Mississippi 39216; and Analytical Chemistry Unit (B.M.), Pathology Queensland, Health Services Support Agency, Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| | - Celso E Gomez-Sanchez
- Cardiovascular and Hormonal Research Laboratory, Department of Cardiology (T.Y.L.L., A.S.M.), Division of Perinatal Research (A.W.A.), Northern Blood Research Centre and Department of Haematology and Transfusion Medicine (M.-C.M.-K.), Sydney Neuro-Oncology Group and Bill Walsh Translational Cancer Research Laboratory (A.H.), Royal North Shore Hospital and Kolling Institute (A.W.A., T.Y.L.L., M.-C.M.-K., A.H., A.S.M.), Royal North Shore Hospital and The University of Sydney, Sydney 2065, Australia; Division of Endocrinology, G. V. (Sonny) Montgomery Veterans Affairs Medical Center and University of Mississippi Medical Center (C.E.G.-S.), Jackson, Mississippi 39216; and Analytical Chemistry Unit (B.M.), Pathology Queensland, Health Services Support Agency, Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| | - Marie-Christine Morel-Kopp
- Cardiovascular and Hormonal Research Laboratory, Department of Cardiology (T.Y.L.L., A.S.M.), Division of Perinatal Research (A.W.A.), Northern Blood Research Centre and Department of Haematology and Transfusion Medicine (M.-C.M.-K.), Sydney Neuro-Oncology Group and Bill Walsh Translational Cancer Research Laboratory (A.H.), Royal North Shore Hospital and Kolling Institute (A.W.A., T.Y.L.L., M.-C.M.-K., A.H., A.S.M.), Royal North Shore Hospital and The University of Sydney, Sydney 2065, Australia; Division of Endocrinology, G. V. (Sonny) Montgomery Veterans Affairs Medical Center and University of Mississippi Medical Center (C.E.G.-S.), Jackson, Mississippi 39216; and Analytical Chemistry Unit (B.M.), Pathology Queensland, Health Services Support Agency, Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| | - Brett McWhinney
- Cardiovascular and Hormonal Research Laboratory, Department of Cardiology (T.Y.L.L., A.S.M.), Division of Perinatal Research (A.W.A.), Northern Blood Research Centre and Department of Haematology and Transfusion Medicine (M.-C.M.-K.), Sydney Neuro-Oncology Group and Bill Walsh Translational Cancer Research Laboratory (A.H.), Royal North Shore Hospital and Kolling Institute (A.W.A., T.Y.L.L., M.-C.M.-K., A.H., A.S.M.), Royal North Shore Hospital and The University of Sydney, Sydney 2065, Australia; Division of Endocrinology, G. V. (Sonny) Montgomery Veterans Affairs Medical Center and University of Mississippi Medical Center (C.E.G.-S.), Jackson, Mississippi 39216; and Analytical Chemistry Unit (B.M.), Pathology Queensland, Health Services Support Agency, Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| | - Amanda Hudson
- Cardiovascular and Hormonal Research Laboratory, Department of Cardiology (T.Y.L.L., A.S.M.), Division of Perinatal Research (A.W.A.), Northern Blood Research Centre and Department of Haematology and Transfusion Medicine (M.-C.M.-K.), Sydney Neuro-Oncology Group and Bill Walsh Translational Cancer Research Laboratory (A.H.), Royal North Shore Hospital and Kolling Institute (A.W.A., T.Y.L.L., M.-C.M.-K., A.H., A.S.M.), Royal North Shore Hospital and The University of Sydney, Sydney 2065, Australia; Division of Endocrinology, G. V. (Sonny) Montgomery Veterans Affairs Medical Center and University of Mississippi Medical Center (C.E.G.-S.), Jackson, Mississippi 39216; and Analytical Chemistry Unit (B.M.), Pathology Queensland, Health Services Support Agency, Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| | - Anastasia S Mihailidou
- Cardiovascular and Hormonal Research Laboratory, Department of Cardiology (T.Y.L.L., A.S.M.), Division of Perinatal Research (A.W.A.), Northern Blood Research Centre and Department of Haematology and Transfusion Medicine (M.-C.M.-K.), Sydney Neuro-Oncology Group and Bill Walsh Translational Cancer Research Laboratory (A.H.), Royal North Shore Hospital and Kolling Institute (A.W.A., T.Y.L.L., M.-C.M.-K., A.H., A.S.M.), Royal North Shore Hospital and The University of Sydney, Sydney 2065, Australia; Division of Endocrinology, G. V. (Sonny) Montgomery Veterans Affairs Medical Center and University of Mississippi Medical Center (C.E.G.-S.), Jackson, Mississippi 39216; and Analytical Chemistry Unit (B.M.), Pathology Queensland, Health Services Support Agency, Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| |
Collapse
|
17
|
Abstract
The clinical impact of cardiovascular disease cannot be underestimated. Equally, the importance of cost-effective management of cardiac failure is a pressing issue in the face of an ageing population and the increasing incidence of metabolic disorders worldwide. Targeting the mineralocorticoid receptor (MR) offers one approach for the treatment of heart failure with current strategies for novel MR therapeutics focusing on harnessing their cardio-protective benefits, but limiting the side effects of existing agents. It is now well accepted that activation of the MR in the cardiovascular system promotes tissue inflammation and fibrosis and has negative consequences for cardiac function and patient outcomes following cardiac events. Indeed, blockade of the MR using one of the two available antagonists (spironolactone and eplerenone) provides significant cardio-protective effects in the clinical and experimental setting. Although the pathways downstream of MR that translate receptor activation into tissue inflammation, fibrosis and dysfunction are still being elucidated, a series of recent studies using cell-selective MR (NR3C2)-null or MR-overexpressing mice have offered many new insights into the role of MR in cardiovascular disease and the control of blood pressure. Dissecting the cell-specific roles of MR signalling in the heart and vasculature to identify those pathways that are critical for MR-dependent responses is an important step towards achieving cardiac-selective therapeutics. The goal of this review is to discuss recent advances in this area that have emerged from the study of tissue-selective MR-null mice, and other targeted transgenic models and their relevance to clinical disease.
Collapse
Affiliation(s)
- Morag J Young
- Cardiovascular EndocrinologyMIMR-PHI Institute, 27-31 Wright St, Clayton 3168, AustraliaDepartment of PhysiologyMonash University, Clayton 3168, Australia Cardiovascular EndocrinologyMIMR-PHI Institute, 27-31 Wright St, Clayton 3168, AustraliaDepartment of PhysiologyMonash University, Clayton 3168, Australia
| | - Amanda J Rickard
- Cardiovascular EndocrinologyMIMR-PHI Institute, 27-31 Wright St, Clayton 3168, AustraliaDepartment of PhysiologyMonash University, Clayton 3168, Australia Cardiovascular EndocrinologyMIMR-PHI Institute, 27-31 Wright St, Clayton 3168, AustraliaDepartment of PhysiologyMonash University, Clayton 3168, Australia
| |
Collapse
|
18
|
Nakagawa H, Oberwinkler H, Nikolaev VO, Gaßner B, Umbenhauer S, Wagner H, Saito Y, Baba HA, Frantz S, Kuhn M. Atrial Natriuretic Peptide Locally Counteracts the Deleterious Effects of Cardiomyocyte Mineralocorticoid Receptor Activation. Circ Heart Fail 2014; 7:814-21. [DOI: 10.1161/circheartfailure.113.000885] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background—
The endocrine balance between atrial natriuretic peptide (ANP) and the renin–angiotensin–aldosterone system is critical for the maintenance of arterial blood pressure and volume homeostasis. This study investigated whether a cardiac imbalance between ANP and aldosterone, toward increased mineralocorticoid receptor (MR) signaling, contributes to adverse left ventricular remodeling in response to pressure overload.
Methods and Results—
We used the MR-selective antagonist eplerenone to test the role of MRs in mediating pressure overload–induced dilatative cardiomyopathy of mice with abolished local, cardiac ANP activity. In response to 21 days of transverse aortic constriction, mice with cardiomyocyte-restricted inactivation (knockout) of the ANP receptor (guanylyl cyclase [GC]-A) or the downstream cGMP-dependent protein kinase I developed enhanced left ventricular hypertrophy and fibrosis together with contractile dysfunction. Treatment with eplerenone (100 mg/kg/d) attenuated left ventricular hypertrophy and fully prevented fibrosis, dilatation, and failure. Transverse aortic constriction induced the cardiac expression of profibrotic connective tissue growth factor and attenuated the expression of SERCA2a (sarcoplasmic reticulum Ca
2+
-ATPase) in knockout mice, but not in controls. These genotype-dependent molecular changes were similarly prevented by eplerenone. ANP attenuated the aldosterone-induced nuclear translocation of MRs via GC-A/cGMP-dependent protein kinase I in transfected HEK 293 (human embryonic kidney) cells. Coimmunoprecipitation and fluorescence resonance energy transfer experiments demonstrated that a population of MRs were membrane associated in close interaction with GC-A and cGMP-dependent protein kinase I and, moreover, that aldosterone caused a conformational change of this membrane MR/GC-A protein complex which was prevented by ANP.
Conclusions—
ANP counter-regulates cardiac MR activation in hypertensive heart disease. An imbalance in cardiac ANP/GC-A (inhibition) and aldosterone/MR signaling (augmentation) favors adverse cardiac remodeling in chronic pressure overload.
Collapse
Affiliation(s)
- Hitoshi Nakagawa
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Heike Oberwinkler
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Viacheslav O. Nikolaev
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Birgit Gaßner
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Sandra Umbenhauer
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Helga Wagner
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Yoshihiko Saito
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Hideo A. Baba
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Stefan Frantz
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Michaela Kuhn
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| |
Collapse
|
19
|
Abstract
The primary adrenal cortical steroid hormones, aldosterone, and the glucocorticoids cortisol and corticosterone, act through the structurally similar mineralocorticoid (MR) and glucocorticoid receptors (GRs). Aldosterone is crucial for fluid, electrolyte, and hemodynamic homeostasis and tissue repair; the significantly more abundant glucocorticoids are indispensable for energy homeostasis, appropriate responses to stress, and limiting inflammation. Steroid receptors initiate gene transcription for proteins that effect their actions as well as rapid non-genomic effects through classical cell signaling pathways. GR and MR are expressed in many tissues types, often in the same cells, where they interact at molecular and functional levels, at times in synergy, others in opposition. Thus the appropriate balance of MR and GR activation is crucial for homeostasis. MR has the same binding affinity for aldosterone, cortisol, and corticosterone. Glucocorticoids activate MR in most tissues at basal levels and GR at stress levels. Inactivation of cortisol and corticosterone by 11β-HSD2 allows aldosterone to activate MR within aldosterone target cells and limits activation of the GR. Under most conditions, 11β-HSD1 acts as a reductase and activates cortisol/corticosterone, amplifying circulating levels. 11β-HSD1 and MR antagonists mitigate inappropriate activation of MR under conditions of oxidative stress that contributes to the pathophysiology of the cardiometabolic syndrome; however, MR antagonists decrease normal MR/GR functional interactions, a particular concern for neurons mediating cognition, memory, and affect.
Collapse
Affiliation(s)
- Elise Gomez-Sanchez
- G.V.(Sonny) Montgomery V.A. Medical Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Celso E. Gomez-Sanchez
- G.V.(Sonny) Montgomery V.A. Medical Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
20
|
Ennis IL, Aiello EA, Cingolani HE, Perez NG. The autocrine/paracrine loop after myocardial stretch: mineralocorticoid receptor activation. Curr Cardiol Rev 2014; 9:230-40. [PMID: 23909633 PMCID: PMC3780348 DOI: 10.2174/1573403x113099990034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 12/13/2012] [Indexed: 01/13/2023] Open
Abstract
The stretch of cardiac muscle increases developed force in two phases. The first phase, which occurs rapidly, constitutes the well-known Frank-Starling mechanism and it is generally attributed to enhanced myofilament responsiveness to Ca(2+). The second phase or slow force response (SFR) occurs gradually and is due to an increase in the calcium transient amplitude as a result of a stretch-triggered autocrine/paracrine mechanism. We previously showed that Ca(2+) entry through reverse Na(+)/Ca(2+) exchange underlies the SFR, as the final step of an autocrine/paracrine cascade involving release of angiotensin II/endothelin, and a Na(+)/H(+) exchanger (NHE-1) activation-mediated rise in Na+. In the present review we mainly focus on our three latest contributions to the understanding of this signalling pathway triggered by myocardial stretch: 1) The finding that an increased production of reactive oxygen species (ROS) from mitochondrial origin is critical in the activation of the NHE-1 and therefore in the genesis of the SFR; 2) the demonstration of a key role played by the transactivation of the epidermal growth factor receptor; and 3) the involvement of mineralocorticoid receptors (MR) activation in the stretch-triggered cascade leading to the SFR. Among these novel contributions, the critical role played by the MR is perhaps the most important one. This finding may conceivably provide a mechanistic explanation to the recently discovered strikingly beneficial effects of MR antagonism in humans with cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Irene L Ennis
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Medicas, Universidad Nacional de La Plata, Argentina
| | | | | | | |
Collapse
|
21
|
Grossmann C, Ruhs S, Langenbruch L, Mildenberger S, Strätz N, Schumann K, Gekle M. Nuclear shuttling precedes dimerization in mineralocorticoid receptor signaling. ACTA ACUST UNITED AC 2014; 19:742-51. [PMID: 22726688 DOI: 10.1016/j.chembiol.2012.04.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 04/22/2012] [Accepted: 04/25/2012] [Indexed: 10/28/2022]
Abstract
The mineralocorticoid receptor (MR), a member of the steroid receptor superfamily, regulates water-electrolyte balance and mediates pathophysiological effects in the renocardiovascular system. Previously, it was assumed that after binding aldosterone, the MR dissociates from HSP90, forms homodimers, and then translocates into the nucleus where it acts as a transcription factor (Guiochon-Mantel et al., 1989; Robertson et al., 1993; Savory et al., 2001). We found that, during aldosterone-induced nuclear translocation, MR is bound to HSP90 both in the cytosol and the nucleus. Homodimerization measured by eBRET and FRET takes place when the MR is already predominantly nuclear. In vitro binding of MR to DNA was independent of ligand but could be partially inhibited by geldanamycin. Overall, here we provide insights into classical MR signaling necessary for elucidating the mechanisms of pathophysiological MR effects and MR specificity.
Collapse
Affiliation(s)
- Claudia Grossmann
- Julius-Bernstein-Institute of Physiology, University Halle-Wittenberg, Halle/Saale 06112, Germany.
| | | | | | | | | | | | | |
Collapse
|
22
|
Wehmeyer L, Du Toit A, Lang DM, Hapgood JP. Lipid raft- and protein kinase C-mediated synergism between glucocorticoid- and gonadotropin-releasing hormone signaling results in decreased cell proliferation. J Biol Chem 2014; 289:10235-51. [PMID: 24558046 DOI: 10.1074/jbc.m113.544742] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cross-talk between the glucocorticoid receptor (GR) and other receptors is emerging as a mechanism for fine-tuning cellular responses. We have previously shown that gonadotropin-releasing hormone (GnRH) ligand-independently activates the GR and synergistically modulates glucocorticoid-induced transcription of an endogenous gene in LβT2 pituitary gonadotrope precursor cells. Here, we investigated GR and GnRH receptor (GnRHR) cross-talk that involves co-localization with lipid rafts in LβT2 cells. We report that the GnRHR and a small population of the GR co-localize with the lipid raft protein flotillin-1 (Flot-1) at the plasma membrane and that the GR is present in a complex with Flot-1, independent of the presence of ligands. We found that the SGK-1 gene is up-regulated by Dex and GnRH alone, whereas a combination of both ligands resulted in a synergistic increase in SGK-1 mRNA levels. Using siRNA-mediated knockdown and antagonist strategies, we show that the gene-specific synergistic transcriptional response requires the GR, GnRHR, and Flot-1 as well as the protein kinase C pathway. Interestingly, although several GR cofactors are differentially recruited to the SGK-1 promoter in the presence of Dex and GnRH, GR levels remain unchanged compared with Dex treatment alone, suggesting that lipid raft association of the GR has a role in enhancing its transcriptional output in the nucleus. Finally, we show that Dex plus GnRH synergistically inhibit cell proliferation in a manner dependent on SGK-1 and Flot-1. Collectively the results support a mechanism whereby GR and GnRHR cross-talk within Flot-1-containing lipid rafts modulates cell proliferation via PKC activation and SGK-1 up-regulation.
Collapse
Affiliation(s)
- Lancelot Wehmeyer
- From the Department of Molecular and Cell Biology, Faculty of Science and
| | | | | | | |
Collapse
|
23
|
De Giusti VC, Ciancio MC, Orlowski A, Aiello EA. Modulation of the cardiac sodium/bicarbonate cotransporter by the renin angiotensin aldosterone system: pathophysiological consequences. Front Physiol 2014; 4:411. [PMID: 24478712 PMCID: PMC3894460 DOI: 10.3389/fphys.2013.00411] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/27/2013] [Indexed: 12/22/2022] Open
Abstract
The sodium/bicarbonate cotransporter (NBC) is one of the major alkalinizing mechanisms in the cardiomyocytes. It has been demonstrated the existence of at least two functional isoforms, one that promotes the co-influx of 1 molecule of Na+ per 1 molecule of HCO−3 (electroneutral isoform; NBCn1) and the other one that generates the co-influx of 1 molecule of Na+ per 2 molecules of HCO−3 (electrogenic isoform; NBCe1). Both isoforms are important to maintain intracellular pH (pHi) and sodium concentration ([Na+]i). In addition, NBCe1 generates an anionic repolarizing current that modulates the action potential duration (APD). The renin-angiotensin-aldosterone system (RAAS) is implicated in the modulation of almost all physiological cardiac functions and is also involved in the development and progression of cardiac diseases. It was reported that angiotensin II (Ang II) exhibits an opposite effect on NBC isoforms: it activates NBCn1 and inhibits NBCe1. The activation of NBCn1 leads to an increase in pHi and [Na+]i, which indirectly, due to the stimulation of reverse mode of the Na+/Ca2+ exchanger (NCX), conduces to an increase in the intracellular Ca2+ concentration. On the other hand, the inhibition of NBCe1 generates an APD prolongation, potentially representing a risk of arrhythmias. In the last years, the potentially altered NBC function in pathological scenarios, as cardiac hypertrophy and ischemia-reperfusion, has raised increasing interest among investigators. This review attempts to draw the attention on the relevant regulation of NBC activity by RAAS, since it modulates pHi and [Na+]i, which are involved in the development of cardiac hypertrophy, the damage produced by ischemia-reperfusion and the generation of arrhythmic events, suggesting a potential role of NBC in cardiac diseases.
Collapse
Affiliation(s)
- Verónica C De Giusti
- Facultad de Ciencias Médicas, Centro de Investigaciones Cardiovasculares, Universidad Nacional de La Plata, CONICET-La Plata La Plata, Argentina
| | - María C Ciancio
- Facultad de Ciencias Médicas, Centro de Investigaciones Cardiovasculares, Universidad Nacional de La Plata, CONICET-La Plata La Plata, Argentina
| | - Alejandro Orlowski
- Facultad de Ciencias Médicas, Centro de Investigaciones Cardiovasculares, Universidad Nacional de La Plata, CONICET-La Plata La Plata, Argentina
| | - Ernesto A Aiello
- Facultad de Ciencias Médicas, Centro de Investigaciones Cardiovasculares, Universidad Nacional de La Plata, CONICET-La Plata La Plata, Argentina
| |
Collapse
|
24
|
Díaz RG, Pérez NG, Morgan PE, Villa-Abrille MC, Caldiz CI, Nolly MB, Portiansky EL, Ennis IL, Cingolani HE. Myocardial mineralocorticoid receptor activation by stretching and its functional consequences. Hypertension 2013; 63:112-8. [PMID: 24126173 DOI: 10.1161/hypertensionaha.113.01726] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Myocardial stretch triggers an angiotensin II-dependent autocrine/paracrine loop of intracellular signals, leading to reactive oxygen species-mediated activation of redox-sensitive kinases. Based on pharmacological strategies, we previously proposed that mineralocorticoid receptor (MR) is necessary for this stretch-triggered mechanism. Now, we aimed to test the role of MR after stretch by using a molecular approach to avoid secondary effects of pharmacological MR blockers. Small hairpin interference RNA capable of specifically knocking down the MR was incorporated into a lentiviral vector (l-shMR) and injected into the left ventricular wall of Wistar rats. The same vector but expressing a nonsilencing sequence (scramble) was used as control. Lentivirus propagation through the left ventricle was evidenced by confocal microscopy. Myocardial MR expression, stretch-triggered activation of redox-sensitive kinases (ERK1/2-p90(RSK)), the consequent Na(+)/H(+) exchanger-mediated changes in pHi (HEPES-buffer), and its mechanical counterpart, the slow force response, were evaluated. Furthermore, reactive oxygen species production in response to a low concentration of angiotensin II (1.0 nmol/L) or an equipotent concentration of epidermal growth factor (0.1 μg/mL) was compared in myocardial tissue slices from both groups. Compared with scramble, animals transduced with l-shMR showed (1) reduced cardiac MR expression, (2) cancellation of angiotensin II-induced reactive oxygen species production but preservation of epidermal growth factor-induced reactive oxygen species production, (3) cancellation of stretch-triggered increase in ERK1/2-p90(RSK) phosphorylation, (4) lack of stretch-induced Na(+)/H(+) exchanger activation, and (5) abolishment of the slow force response. Our results provide strong evidence that MR activation occurs after myocardial stretch and is a key factor to promote redox-sensitive kinase activation and their downstream consequences.
Collapse
Affiliation(s)
- Romina G Díaz
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, UNLP, 60 y 120, 1900 La Plata, Argentina.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Aldosterone is now recognized as an increasingly important contributor to cardiometabolic pathology via inflammatory and fibrosis-related pathways in addition to its classically described role in sodium and volume regulation. Consequently, much effort has been directed towards characterizing the molecular pathways involved in aldosterone-mediated fibrosis and inflammation. What was once viewed as straightforward steroid hormone biology is now appreciated as a highly complex and tightly regulated series of pathways and interactions. These recognitions have fuelled a multidisciplinary effort to identify precisely how aldosterone mediates intracellular activation of both genomic (latent) and nongenomic (rapid) mechanisms of influence. This review will explore recent novel pathways regulating aldosterone action, focusing on the nongenomic pathways. RECENT FINDINGS Several recent discoveries have redefined our understanding of aldosterone interactions at the cellular level. This includes activation of the mineralocorticoid receptor at the plasma membrane instead of via classical nuclear hormone receptor interaction, and identification of novel cofactor scaffolding proteins that modify aldosterone influence at the cellular level. In addition, aldosterone activation of secondary messenger system cascades can occur directly and independent of mineralocorticoid receptor interaction. SUMMARY Substantial progress in detailing the molecular biology of aldosterone regulation and action should facilitate study of how it exerts detrimental effects in cardiometabolic diseases. However, to date, the clinical impact of these discoveries has not been validated. Translational efforts are now required to determine if novel therapeutic targets can be developed.
Collapse
Affiliation(s)
- Jonathan S Williams
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital/Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
26
|
De Giusti VC, Caldiz CI, Ennis IL, Pérez NG, Cingolani HE, Aiello EA. Mitochondrial reactive oxygen species (ROS) as signaling molecules of intracellular pathways triggered by the cardiac renin-angiotensin II-aldosterone system (RAAS). Front Physiol 2013; 4:126. [PMID: 23755021 PMCID: PMC3667248 DOI: 10.3389/fphys.2013.00126] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 05/13/2013] [Indexed: 12/22/2022] Open
Abstract
Mitochondria represent major sources of basal reactive oxygen species (ROS) production of the cardiomyocyte. The role of ROS as signaling molecules that mediate different intracellular pathways has gained increasing interest among physiologists in the last years. In our lab, we have been studying the participation of mitochondrial ROS in the intracellular pathways triggered by the renin-angiotensin II-aldosterone system (RAAS) in the myocardium during the past few years. We have demonstrated that acute activation of cardiac RAAS induces mitochondrial ATP-dependent potassium channel (mitoKATP) opening with the consequent enhanced production of mitochondrial ROS. These oxidant molecules, in turn, activate membrane transporters, as sodium/hydrogen exchanger (NHE-1) and sodium/bicarbonate cotransporter (NBC) via the stimulation of the ROS-sensitive MAPK cascade. The stimulation of such effectors leads to an increase in cardiac contractility. In addition, it is feasible to suggest that a sustained enhanced production of mitochondrial ROS induced by chronic cardiac RAAS, and hence, chronic NHE-1 and NBC stimulation, would also result in the development of cardiac hypertrophy.
Collapse
Affiliation(s)
- V C De Giusti
- Facultad de Ciencias Médicas, Centro de Investigaciones Cardiovasculares, UNLP-CONICET La Plata, Argentina
| | | | | | | | | | | |
Collapse
|
27
|
Martinerie L, Munier M, Le Menuet D, Meduri G, Viengchareun S, Lombès M. The mineralocorticoid signaling pathway throughout development: expression, regulation and pathophysiological implications. Biochimie 2012; 95:148-57. [PMID: 23026756 DOI: 10.1016/j.biochi.2012.09.030] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Accepted: 09/23/2012] [Indexed: 10/27/2022]
Abstract
The mineralocorticoid signaling pathway has gained interest over the past few years, considering not only its implication in numerous pathologies but also its emerging role in physiological processes during kidney, brain, heart and lung development. This review aims at describing the setting and regulation of aldosterone biosynthesis and the expression of the mineralocorticoid receptor (MR), a nuclear receptor mediating aldosterone action in target tissues, during the perinatal period. Specificities concerning MR expression and regulation during the development of several major organs are highlighted. We provide evidence that MR expression is tightly controlled in a tissue-specific manner during development, which could have major pathophysiological implications in the neonatal period.
Collapse
|
28
|
Wendler A, Albrecht C, Wehling M. Nongenomic actions of aldosterone and progesterone revisited. Steroids 2012; 77:1002-6. [PMID: 22285849 DOI: 10.1016/j.steroids.2011.12.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 12/20/2011] [Indexed: 01/16/2023]
Abstract
After almost 30 years of research, the existence of nongenomic steroid actions is no longer disputed. Yet, there is still a debate on the nature of receptors involved, and answers to the inherent questions are important for translational activities. In the case of aldosterone, the existence of receptors different from the classic mineralocorticoid receptors (MR) had been postulated 25 years ago as the pharmacology of about 50% of rapid actions of aldosterone reported so far is incompatible with MR involvement (insensitivity to classic MR antagonists). Candidates proposed as alternatives to MR were protein kinase C, sodium-potassium ATPase or aberrant forms of MR, none of which supported convincing evidence to represent 'the aldosterone membrane receptor'. Early in 2011, data on GPR30 showed its involvement in rapid aldosterone action, and major pharmacological aspects of this action are compatible with the landmark deviations from MR pharmacology mentioned above. GPR30, therefore, may be a receptor candidate for nongenomic aldosterone action. Similarly, a variety of promising candidates mediating rapid progesterone action has been described, including progesterone receptor membrane component 1 (PGRMC1) which seems to be associated with tumor proliferation, and membrane progesterone receptor (mPR) originally identified in fish with potential linkage to reproductive processes. So far, no candidate was unanimously convincing. In 2010, two independent groups reported that CatSper, a calcium channel, is a strong receptor candidate for the rapid action of progesterone on sperm fertilization. With these novel receptors cloned, translational activities ultimately leading to new drugs for cardiovascular protection (in the case of aldosterone) or fertilization benefits (for progesterone) are much more promising.
Collapse
Affiliation(s)
- Alexandra Wendler
- University of Heidelberg, Clinical Pharmacology Mannheim, Maybachstr. 14, D-68169 Mannheim, Germany
| | | | | |
Collapse
|
29
|
Caldiz CI, Díaz RG, Nolly MB, Chiappe de Cingolani GE, Ennis IL, Cingolani HE, Pérez NG. Mineralocorticoid receptor activation is crucial in the signalling pathway leading to the Anrep effect. J Physiol 2012; 589:6051-61. [PMID: 22174146 DOI: 10.1113/jphysiol.2011.218750] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The increase in myocardial reactive oxygen species after epidermal growth factor receptor transactivation is a crucial step in the autocrine/paracrine angiotensin II/endothelin receptor activation leading to the slow force response to stretch (SFR). Since experimental evidence suggests a link between angiotensin II or its AT1 receptor and the mineralocorticoid receptor (MR), and MR transactivates the epidermal growth factor receptor, we thought to determine whether MR activation participates in the SFR development in rat myocardium. We show here that MR activation is necessary to promote reactive oxygen species formation by a physiological concentration of angiotensin II (1 nmol l(-1)), since an increase in superoxide anion formation of ~50% of basal was suppressed by blocking MR with spironolactone or eplerenone. This effect was also suppressed by blocking AT1, endothelin (type A) or epidermal growth factor receptors, by inhibiting NADPH oxydase or by targeting mitochondria, and was unaffected by glucocorticoid receptor inhibition. All interventions except AT1 receptor blockade blunted the increase in superoxide anion promoted by an equipotent dose of endothelin-1 (1 nmol l(-1)) confirming that endothelin receptors activation is downstream of AT1. Similarly, an increase in superoxide anion promoted by an equipotent dose of aldosterone (10 nmol l(-1)) was blocked by spironolactone or eplerenone, by preventing epidermal growth factor receptor transactivation, but not by inhibiting glucocorticoid receptors or protein synthesis, suggesting non-genomic MR effects. Combination of aldosterone plus endothelin-1 did not increase superoxide anion formation more than each agonist separately. We found that aldosterone increased phosphorylation of the redox-sensitive kinases ERK1/2-p90RSK and the NHE-1, effects that were eliminated by eplerenone or by preventing epidermal growth factor receptor transactivation. Finally, we provide evidence that the SFR is suppressed by MR blockade, by preventing epidermal growth factor receptor transactivation or by scavenging reactive oxygen species, but it is unaffected by glucocorticoid receptor blockade or protein synthesis inhibition. Our results suggest that MR activation is a necessary step in the stretch-triggered reactive oxygen species-mediated activation of redox-sensitive kinases upstream NHE-1.
Collapse
Affiliation(s)
- Claudia I Caldiz
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | | | | | | | | | | | | |
Collapse
|
30
|
Grossmann C, Gekle M. Interaction between mineralocorticoid receptor and epidermal growth factor receptor signaling. Mol Cell Endocrinol 2012; 350:235-41. [PMID: 21827828 DOI: 10.1016/j.mce.2011.07.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 07/22/2011] [Accepted: 07/23/2011] [Indexed: 02/05/2023]
Abstract
The mineralocorticoid receptor (MR) is a steroid receptor that physiologically regulates water and electrolyte homeostasis but that can also induce pathophysiological effects in the renocardiovascular system. Classically, the MR acts as a transcription factor at glucocorticoid response elements but additional protein-protein interactions with other signaling cascades have been described. Of these, the crosstalk with EGFR signaling is especially interesting because various vasoactive substances like angiotensin II and endothelin-1 also mediate their pathophysiological effects via the EGFR. Recently, the MR has been shown to interact nongenomically (via transactivation) and genomically with the epidermal growth factor receptor (via altered expression). These interactions seem to contribute to physiological (e.g. salt homeostasis) as well as pathophysiological (e.g. vascular function) MR effects. The current knowledge on the mechanisms of interaction and on the possible cellular and systemic physiological as well as pathophysiological relevance is reviewed in this article.
Collapse
Affiliation(s)
- Claudia Grossmann
- Julius-Bernstein-Institute of Physiology, University of Halle-Wittenberg, Magdeburger Strasse 6, Halle, Germany.
| | | |
Collapse
|
31
|
Groeneweg FL, Karst H, de Kloet ER, Joëls M. Mineralocorticoid and glucocorticoid receptors at the neuronal membrane, regulators of nongenomic corticosteroid signalling. Mol Cell Endocrinol 2012; 350:299-309. [PMID: 21736918 DOI: 10.1016/j.mce.2011.06.020] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 06/15/2011] [Accepted: 06/20/2011] [Indexed: 02/06/2023]
Abstract
The balance between corticosteroid actions induced via activation of the mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR) determines the brain's response to stress. While both receptors are best known for their delayed genomic role, it has become increasingly evident that they can also associate with the plasma membrane and act as mediators of rapid, nongenomic signalling. Nongenomic corticosteroid actions in the brain are required for the coordination of a rapid adaptive response to stress; membrane-associated MRs and GRs play a major role herein. However, many questions regarding the underlying mechanism are still unresolved. How do MR and GR translocate to the membrane and what are their downstream signalling partners? In this review we discuss these issues based on insights obtained from related receptors, most notably the estrogen receptor α.
Collapse
Affiliation(s)
- Femke L Groeneweg
- Department of Medical Pharmacology, Leiden Amsterdam Centre for Drug Research, Leiden University Medical Centre, Leiden University, Einsteinweg 55, Leiden, The Netherlands.
| | | | | | | |
Collapse
|
32
|
Dooley R, Harvey BJ, Thomas W. Non-genomic actions of aldosterone: from receptors and signals to membrane targets. Mol Cell Endocrinol 2012; 350:223-34. [PMID: 21801805 DOI: 10.1016/j.mce.2011.07.019] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 07/05/2011] [Accepted: 07/09/2011] [Indexed: 10/17/2022]
Abstract
In tissues which express the mineralocorticoid receptor (MR), aldosterone modulates the expression of membrane targets such as the subunits of the epithelial Na(+) channel, in combination with important signalling intermediates such as serum and glucocorticoid-regulated kinase-1. In addition, the rapid 'non-genomic' activation of protein kinases and secondary messenger signalling cascades has also been detected in aldosterone-sensitive tissues of the nephron, distal colon and cardiovascular system. These rapid actions are variously described as being coupled to MR or to an as yet unidentified, membrane-associated aldosterone receptor. The rapidly activated signalling cascades add a level of fine-tuning to the activity of aldosterone-responsive membrane transporters and also modulate the aldosterone-induced changes in gene expression through receptor and transcription factor phosphorylation.
Collapse
Affiliation(s)
- Ruth Dooley
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland
| | | | | |
Collapse
|
33
|
Gomez-Sanchez CE, Warden M, Gomez-Sanchez MT, Hou X, Gomez-Sanchez EP. Diverse immunostaining patterns of mineralocorticoid receptor monoclonal antibodies. Steroids 2011; 76:1541-5. [PMID: 21945398 PMCID: PMC3217130 DOI: 10.1016/j.steroids.2011.09.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 09/04/2011] [Accepted: 09/06/2011] [Indexed: 12/21/2022]
Abstract
The mineralocorticoid receptor (MR) is a widely distributed ligand activated nuclear transcription factor that is bound by various chaperone proteins that alter its conformation depending upon its location in the cell and whether it is ligand-bound. We describe the development and characterization of new monoclonal antibodies produced against a rat recombinant protein corresponding to aminoacids 5-550 of the MR to produce antibodies that recognize the receptor in specific conformations. Most of the resulting monoclonal antibodies studied were similar to those we produced by immunization with peptide isotopes, however two detected a single band at the appropriate molecular mass as the MR and had distinct immunostaining characteristics in neurons. One labeled cytosolic MR, the other labeled membranes and cytosol, including axons. These antibodies will permit study of the subcellular localization of the MR under various physiological and pathological conditions. We have also confirmed that the MR is highly unstable and requires special handling.
Collapse
Affiliation(s)
- Celso E Gomez-Sanchez
- Research Service, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS 39110, USA.
| | | | | | | | | |
Collapse
|
34
|
Walczak C, Gaignier F, Gilet A, Zou F, Thornton SN, Ropars A. Aldosterone increases VEGF-A production in human neutrophils through PI3K, ERK1/2 and p38 pathways. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:2125-32. [PMID: 21803079 DOI: 10.1016/j.bbamcr.2011.07.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 07/15/2011] [Accepted: 07/18/2011] [Indexed: 01/20/2023]
Abstract
Aldosterone is now recognised as an important actor in inflammation processes. Neoangiogenesis plays a crucial role in this complex process and immune cells, such as neutrophils, appear to be able to secrete different forms of (pro)angiogenic molecules, especially VEGF-A. The present work was undertaken to investigate whether aldosterone was able to regulate VEGF-A production in human neutrophils. The HL-60 (progranulocytic) cell line and human polymorphonuclear leukocytes were incubated for different time periods with aldosterone. Total cellular RNA extraction, submitted to reverse transcription and real time semi-quantitative PCR, was used to study VEGF-A mRNA expression. Cell supernatants were collected and ELISA tests were performed to analyse VEGF-A protein production. Aldosterone increased VEGF-A mRNA and protein expression in a dose- and time-dependent manner in both cell types. Inhibitors of PI3 kinases, ERK1/2, and to a lesser extent of p38 MAPK, decreased this aldosterone-induced immune cell activation. Western-blot performed with HL-60 cells confirmed that ERK1/2 and p38 MAPK pathways were stimulated by aldosterone. Mineralocorticoid receptors are implicated in this VEGF-A up-regulation because HL-60 cells pre-treated with spironolactone, an aldosterone receptor antagonist, diminished the effects of aldosterone. Aldosterone was also able to increase VEGF-A production of phagocytic cells such as neutrophils. These results suggest that this hormone could play an active role in the neovascularisation process by favouring entry of plasma proteins and fluids into the vascular wall, cell proliferation and tissue rebuilding.
Collapse
Affiliation(s)
- Cécile Walczak
- University of Henri Poincaré, Vandoeuvre-les-Nacy, France
| | | | | | | | | | | |
Collapse
|
35
|
Krug AW, Pojoga LH, Williams GH, Adler GK. Cell Membrane–Associated Mineralocorticoid Receptors? Hypertension 2011; 57:1019-25. [DOI: 10.1161/hypertensionaha.110.159459] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Alexander W. Krug
- From the Brigham and Women's Hospital/Harvard Medical School, Department of Endocrinology, Diabetes, and Hypertension, Boston, MA
| | - Luminita H. Pojoga
- From the Brigham and Women's Hospital/Harvard Medical School, Department of Endocrinology, Diabetes, and Hypertension, Boston, MA
| | - Gordon H. Williams
- From the Brigham and Women's Hospital/Harvard Medical School, Department of Endocrinology, Diabetes, and Hypertension, Boston, MA
| | - Gail K. Adler
- From the Brigham and Women's Hospital/Harvard Medical School, Department of Endocrinology, Diabetes, and Hypertension, Boston, MA
| |
Collapse
|