1
|
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
|
2
|
Stücheli S, Araya S, Ercan C, Moser SO, Gallon J, Jenö P, Piscuoglio S, Terracciano L, Odermatt A. The Potential Tumor-Suppressor DHRS7 Inversely Correlates with EGFR Expression in Prostate Cancer Cells and Tumor Samples. Cancers (Basel) 2022; 14:cancers14133074. [PMID: 35804847 PMCID: PMC9264982 DOI: 10.3390/cancers14133074] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Prostate cancer is one of the most common malignancies in men. Current therapies are initially effective but resistance often develops, leading to tumor recurrence and death. Further research on new players, mechanisms involved in prostate cancer, and therapy resistance is needed. We studied the role of DHRS7, a potential tumor suppressor with currently unknown physiological function, in prostate cancer cells using proteome and gene expression analyses. Despite the fact that DHRS7 can inactivate 5α-dihydrotestosterone, its effect on prostate cancer cells seems to be unrelated to androgen metabolism. When comparing three widely studied prostate cancer cell lines, we observed a negative correlation between DHRS7 and EGFR expression. DHRS7 knockdown enhanced EGFR expression, while knockdown of EGFR tended to increase DHRS7 expression. Importantly, DHRS7 expression negatively correlates with EGFR expression and positively with survival rates in prostate cancer patients. This study suggests a tumor-suppressor role for DHRS7 by modulating EGFR expression in prostate cancer. Abstract Prostate cancer (PCa), one of the most common malignancies in men, typically responds to initial treatment, but resistance to therapy often leads to metastases and death. The dehydrogenase/reductase 7 (DHRS7, SDR34C1) is an “orphan” enzyme without known physiological function. DHRS7 was previously found to be decreased in higher-stage PCa, and siRNA-mediated knockdown increased the aggressiveness of LNCaP cells. To further explore the role of DHRS7 in PCa, we analyzed the proteome of LNCaP cells following DHRS7 knockdown to assess potentially altered pathways. Although DHRS7 is able to inactivate 5α-dihydrotestosterone, DHRS7 knockdown did not affect androgen receptor (AR) target gene expression, and its effect on PCa cells seems to be androgen-independent. Importantly, proteome analyses revealed increased expression of epidermal growth factor receptor (EGFR), which was confirmed by RT-qPCR and Western blotting. Comparison of AR-positive LNCaP with AR-negative PC-3 and DU145 PCa cell lines revealed a negative correlation between DHRS7 and EGFR expression. Conversely, EGFR knockdown enhanced DHRS7 expression in these cells. Importantly, analysis of patient samples revealed a negative correlation between DHRS7 and EGFR expression, both at the mRNA and protein levels, and DHRS7 expression correlated positively with patient survival rates. These results suggest a protective role for DHRS7 in PCa.
Collapse
Affiliation(s)
- Simon Stücheli
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (S.S.); (S.A.); (S.O.M.)
| | - Selene Araya
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (S.S.); (S.A.); (S.O.M.)
| | - Caner Ercan
- Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland; (C.E.); (S.P.); (L.T.)
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland;
| | - Seraina O. Moser
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (S.S.); (S.A.); (S.O.M.)
| | - John Gallon
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland;
| | - Paul Jenö
- Proteomics Core Facility, Biozentrum, University of Basel, 4056 Basel, Switzerland;
| | - Salvatore Piscuoglio
- Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland; (C.E.); (S.P.); (L.T.)
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland;
| | - Luigi Terracciano
- Institute of Medical Genetics and Pathology, University Hospital Basel, 4031 Basel, Switzerland; (C.E.); (S.P.); (L.T.)
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (S.S.); (S.A.); (S.O.M.)
- Correspondence: ; Tel.: +41-61-207-15-30
| |
Collapse
|
3
|
Lin X, Ullah MHE, Wu X, Xu F, Shan SK, Lei LM, Yuan LQ, Liu J. Cerebro-Cardiovascular Risk, Target Organ Damage, and Treatment Outcomes in Primary Aldosteronism. Front Cardiovasc Med 2022; 8:798364. [PMID: 35187110 PMCID: PMC8847442 DOI: 10.3389/fcvm.2021.798364] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/20/2021] [Indexed: 02/03/2023] Open
Abstract
Primary aldosteronism (PA) is the most common type of endocrine hypertension, and numerous experimental and clinical evidence have verified that prolonged exposure to excess aldosterone is responsible for an increased risk of cerebro-cardiovascular events and target organ damage (TOD) in patients with PA. Therefore, focusing on restoring the toxic effects of excess aldosterone on the target organs is very important to reduce cerebro-cardiovascular events. Current evidence convincingly demonstrates that both surgical and medical treatment strategies would benefit cerebro-cardiovascular outcomes and mortality in the long term. Understanding cerebro-cardiovascular risk in PA would help clinical doctors to achieve both early diagnosis and treatment. Therefore, in this review, we will summarize the cerebro-cardiovascular risk in PA, focusing on the TOD of aldosterone, including brain, heart, vascular system, renal, adipose tissues, diabetes, and obstructive sleep apnea (OSA). Furthermore, the various treatment outcomes of adrenalectomy and medical treatment for patients with PA will also be discussed. We hope this knowledge will help improve cerebro-cardiovascular prognosis and reduce the incidence and mortality of cerebro-cardiovascular events in patients with PA.
Collapse
Affiliation(s)
- Xiao Lin
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Muhammad Hasnain Ehsan Ullah
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiong Wu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Xu
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Su-Kang Shan
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li-Min Lei
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Qing Yuan
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
- Ling-Qing Yuan
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Medical Imaging in Hunan Province, Changsha, China
- Department of Radiology Quality Control Center in Hunan Province, Changsha, China
- *Correspondence: Jun Liu
| |
Collapse
|
4
|
Abstract
Neurohormones and inflammatory mediators have effects in both the heart and the peripheral vasculature. In patients with heart failure (HF), neurohormonal activation and increased levels of inflammatory mediators promote ventricular remodeling and development of HF, as well as vascular dysfunction and arterial stiffness. These processes may lead to a vicious cycle, whereby arterial stiffness perpetuates further ventricular remodeling leading to exacerbation of symptoms. Although significant advances have been made in the treatment of HF, currently available treatment strategies slow, but do not halt, this cycle. The current treatment for HF patients involves the inhibition of neurohormonal activation, which can reduce morbidity and mortality related to this condition. Beyond benefits associated with neurohormonal blockade, other strategies have focused on inhibition of inflammatory pathways implicated in the pathogenesis of HF. Unfortunately, attempts to target inflammation have not yet been successful to improve prognosis of HF. Further work is required to interrupt key maladaptive mechanisms involved in disease progression.
Collapse
|
5
|
Stroedecke K, Meinel S, Markwardt F, Kloeckner U, Straetz N, Quarch K, Schreier B, Kopf M, Gekle M, Grossmann C. The mineralocorticoid receptor leads to increased expression of EGFR and T-type calcium channels that support HL-1 cell hypertrophy. Sci Rep 2021; 11:13229. [PMID: 34168192 PMCID: PMC8225817 DOI: 10.1038/s41598-021-92284-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/05/2021] [Indexed: 11/12/2022] Open
Abstract
The EGF receptor (EGFR) has been extensively studied in tumor biology and recently a role in cardiovascular pathophysiology was suggested. The mineralocorticoid receptor (MR) is an important effector of the renin-angiotensin-aldosterone-system and elicits pathophysiological effects in the cardiovascular system; however, the underlying molecular mechanisms are unclear. Our aim was to investigate the importance of EGFR for MR-mediated cardiovascular pathophysiology because MR is known to induce EGFR expression. We identified a SNP within the EGFR promoter that modulates MR-induced EGFR expression. In RNA-sequencing and qPCR experiments in heart tissue of EGFR KO and WT mice, changes in EGFR abundance led to differential expression of cardiac ion channels, especially of the T-type calcium channel CACNA1H. Accordingly, CACNA1H expression was increased in WT mice after in vivo MR activation by aldosterone but not in respective EGFR KO mice. Aldosterone- and EGF-responsiveness of CACNA1H expression was confirmed in HL-1 cells by Western blot and by measuring peak current density of T-type calcium channels. Aldosterone-induced CACNA1H protein expression could be abrogated by the EGFR inhibitor AG1478. Furthermore, inhibition of T-type calcium channels with mibefradil or ML218 reduced diameter, volume and BNP levels in HL-1 cells. In conclusion the MR regulates EGFR and CACNA1H expression, which has an effect on HL-1 cell diameter, and the extent of this regulation seems to depend on the SNP-216 (G/T) genotype. This suggests that the EGFR may be an intermediate for MR-mediated cardiovascular changes and that SNP analysis can help identify subgroups of patients that will benefit most from MR antagonists.
Collapse
Affiliation(s)
- Katharina Stroedecke
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06097, Halle, Saale, Germany
| | - Sandra Meinel
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06097, Halle, Saale, Germany
| | - Fritz Markwardt
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06097, Halle, Saale, Germany
| | - Udo Kloeckner
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06097, Halle, Saale, Germany
| | - Nicole Straetz
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06097, Halle, Saale, Germany
| | - Katja Quarch
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06097, Halle, Saale, Germany
| | - Barbara Schreier
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06097, Halle, Saale, Germany
| | - Michael Kopf
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06097, Halle, Saale, Germany
| | - Michael Gekle
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06097, Halle, Saale, Germany
| | - Claudia Grossmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06097, Halle, Saale, Germany.
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Spencer S, Wheeler‐Jones C, Elliott J. Aldosterone and the mineralocorticoid receptor in renal injury: A potential therapeutic target in feline chronic kidney disease. J Vet Pharmacol Ther 2020; 43:243-267. [PMID: 32128854 PMCID: PMC8614124 DOI: 10.1111/jvp.12848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/20/2020] [Accepted: 02/09/2020] [Indexed: 12/24/2022]
Abstract
There is a growing body of experimental and clinical evidence supporting mineralocorticoid receptor (MR) activation as a powerful mediator of renal damage in laboratory animals and humans. Multiple pathophysiological mechanisms are proposed, with the strongest evidence supporting aldosterone-induced vasculopathy, exacerbation of oxidative stress and inflammation, and increased growth factor signalling promoting fibroblast proliferation and deranged extracellular matrix homeostasis. Further involvement of the MR is supported by extensive animal model experiments where MR antagonists (such as spironolactone and eplerenone) abrogate renal injury, including ischaemia-induced damage. Additionally, clinical trials have shown MR antagonists to be beneficial in human chronic kidney disease (CKD) in terms of reducing proteinuria and cardiovascular events, though current studies have not evaluated primary end points which allow conclusions to made about whether MR antagonists reduce mortality or slow CKD progression. Although differences between human and feline CKD exist, feline CKD shares many characteristics with human disease including tubulointerstitial fibrosis. This review evaluates the evidence for the role of the MR in renal injury and summarizes the literature concerning aldosterone in feline CKD. MR antagonists may represent a promising therapeutic strategy in feline CKD.
Collapse
Affiliation(s)
- Sarah Spencer
- Comparative Biomedical SciencesThe Royal Veterinary CollegeLondonUK
| | | | - Jonathan Elliott
- Comparative Biomedical SciencesThe Royal Veterinary CollegeLondonUK
| |
Collapse
|
9
|
De Pascale MR, Della Mura N, Vacca M, Napoli C. Useful applications of growth factors for cardiovascular regenerative medicine. Growth Factors 2020; 38:35-63. [PMID: 33028111 DOI: 10.1080/08977194.2020.1825410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Novel advances for cardiovascular diseases (CVDs) include regenerative approaches for fibrosis, hypertrophy, and neoangiogenesis. Studies indicate that growth factor (GF) signaling could promote heart repair since most of the evidence is derived from preclinical models. Observational studies have evaluated GF serum/plasma levels as feasible biomarkers for risk stratification of CVDs. Noteworthy, two clinical interventional published studies showed that the administration of growth factors (GFs) induced beneficial effect on left ventricular ejection fraction (LVEF), myocardial perfusion, end-systolic volume index (ESVI). To date, large scale ongoing studies are in Phase I-II and mostly focussed on intramyocardial (IM), intracoronary (IC) or intravenous (IV) administration of vascular endothelial growth factor (VEGF) and fibroblast growth factor-23 (FGF-23) which result in the most investigated GFs in the last 10 years. Future data of ongoing randomized controlled studies will be crucial in understanding whether GF-based protocols could be in a concrete way effective in the clinical setting.
Collapse
Affiliation(s)
| | | | - Michele Vacca
- Division of Immunohematology and Transfusion Medicine, Cardarelli Hospital, Naples, Italy
| | - Claudio Napoli
- IRCCS Foundation SDN, Naples, Italy
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
10
|
Barrera-Chimal J, Girerd S, Jaisser F. Mineralocorticoid receptor antagonists and kidney diseases: pathophysiological basis. Kidney Int 2019; 96:302-319. [PMID: 31133455 DOI: 10.1016/j.kint.2019.02.030] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/04/2019] [Accepted: 02/20/2019] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) represents a global health concern, and its prevalence is increasing. The ultimate therapeutic option for CKD is kidney transplantation. However, the use of drugs that target specific pathways to delay or halt CKD progression, such as angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, and sodium-glucose co-transporter-2 (SGLT-2) inhibitors is limited in clinical practice. Mineralocorticoid receptor activation in nonclassical tissues, such as the endothelium, smooth muscle cells, inflammatory cells, podocytes, and fibroblasts may have deleterious effects on kidney structure and function. Several preclinical studies have shown that mineralocorticoid receptor antagonists (MRAs) ameliorate or cure kidney injury and dysfunction in different models of kidney disease. In this review, we present the preclinical evidence showing a benefit of MRAs in acute kidney injury, the transition from acute kidney injury to CKD, hypertensive and diabetic nephropathy, glomerulonephritis, and kidney toxicity induced by calcineurin inhibitors. We also discuss the molecular mechanisms responsible for renoprotection related to MRAs that lead to reduced oxidative stress, inflammation, fibrosis, and hemodynamic alterations. The available clinical data support a benefit of MRA in reducing proteinuria in diabetic kidney disease and improving cardiovascular outcomes in CKD patients. Moreover, a benefit of MRAs in kidney transplantation has also been observed. The past and present clinical trials describing the effect of MRAs on kidney injury are presented, and the risk of hyperkalemia and use of other options, such as potassium binding agents or nonsteroidal MRAs, are also addressed. Altogether, the available preclinical and clinical data support a benefit of using MRAs in CKD, an approach that should be further explored in future clinical trials.
Collapse
Affiliation(s)
- Jonatan Barrera-Chimal
- Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Medicina Traslacional, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Sophie Girerd
- Transplant Unit, Nephrology Department, Nancy University Hospital, Lorraine University, Vandoeuvre-lès-Nancy, France; Institut national de la santé et de la recherche médicale U1116, Clinical Investigation Centre, Lorraine University, Vandoeuvre-lès-Nancy, France; Investigation Network Initiative - Cardiovascular and Renal Clinical Trialists, French-Clinical Research Infrastructure Network, Nancy, France
| | - Frederic Jaisser
- Institut national de la santé et de la recherche médicale U1116, Clinical Investigation Centre, Lorraine University, Vandoeuvre-lès-Nancy, France; Investigation Network Initiative - Cardiovascular and Renal Clinical Trialists, French-Clinical Research Infrastructure Network, Nancy, France; Institut national de la santé et de la recherche médicale, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Sorbonne University, Paris Descartes University, Paris, France.
| |
Collapse
|
11
|
Le-Rademacher JG, Rowland K, Atherton PJ, Dakhil C, Sun Z, Tan A, Schmidt L, Nguyen PL, Radecki Breitkopf C, Pittelkow M, Tindall D, Menon S, Jatoi A. Androgen Mediation-and Antiandrogens Mitigation-of the Epidermal Growth Factor Receptor (EGFR) Inhibitor-Induced Rash: Results From a Pilot Randomized Trial and Small Translational Case Series. Am J Hosp Palliat Care 2019; 36:519-525. [PMID: 30602311 DOI: 10.1177/1049909118819820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Although 50% to 90% of patients who receive epidermal growth factor receptor (EGFR) inhibitors develop a rash, options for rash prevention or palliation remain limited. This issue is particularly important from a palliative care standpoint because these agents are prescribed only to patients with incurable cancer. Here, we report (1) gene expression profiling of skin biopsies from patients with an EGFR inhibitor-induced rash and (2) a randomized, placebo-controlled feasibility trial with the antiandrogen, spironolactone. Both investigations were undertaken to begin to explore the hypothesis that androgens mediate EGFR inhibitor-induced rash and that antiandrogens palliate it. METHODS/RESULTS First, 4 skin biopsies from patients with EGFR inhibitor-induced rash (3 men and 1 woman) were subject to gene expression microarray profiling. A public data set of normal skin gene expression (Gene Expression Omnibus, GSE22998) served as a reference. Sixty percent of commonly interrogated androgen receptor genes (207 of 308 between the 2 data sets) were differentially expressed ( P < .05) in the rash samples. Second, in a 17-patient double-blinded, placebo-controlled trial with topical spironolactone applied to the face, although the primary feasibility end point was not achieved, patients in the spironolactone arm received more doses of EGFR inhibitor, and anecdotal photographic evidence suggested salutatory effects of spironolactone on rash. CONCLUSIONS Epidermal growth factor receptor inhibitor-induced rash appears to be androgen-mediated; antiandrogen therapy merits further study for rash prevention/palliation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Smitha Menon
- 5 Medical College of Wisconsin, Milwaukee, WI, USA
| | | |
Collapse
|
12
|
Schreier B, Wolf A, Hammer S, Pohl S, Mildenberger S, Rabe S, Gekle M, Zipprich A. The selective mineralocorticoid receptor antagonist eplerenone prevents decompensation of the liver in cirrhosis. Br J Pharmacol 2018; 175:2956-2967. [PMID: 29682743 DOI: 10.1111/bph.14341] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/27/2018] [Accepted: 03/08/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE The mineralocorticoid receptor (MR) contributes to fibrosis in various tissues, and MR antagonists, like eplerenone, are used to prevent fibrosis. The role of MR antagonists in hepatic fibrosis and cirrhosis is unknown. Here, we investigated the role of MRs and eplerenone in cirrhosis development. EXPERIMENTAL APPROACH Liver fibrosis (5 weeks) and cirrhosis, without (8 weeks) and with ascites (12 weeks), were induced by CCl4 in rats and comprehensively analysed. The effect of eplerenone on the development of cirrhosis with ascites was assessed. MR expression, cellular and subcellular distribution and impact of hypoxia were investigated in vivo and ex vivo. Primary rat hepatocytes and cell lines were used to investigate MR trafficking and transcriptional activity mechanistically. KEY RESULTS In cirrhosis with ascites, MR mRNA and protein expressions were reduced in hepatocytes of hypoxic areas. While in normoxic areas MRs were mainly cytosolic, the remaining MRs in hypoxic areas were mainly localized in the nuclei, indicating activation followed by translocation and degradation. Accordingly, eplerenone treatment prevented nuclear MR translocation and the worsening of cirrhosis. Exposing hepatocytes ex vivo to hypoxia induced nuclear MR translocation and enhanced transcriptional MR activity at response elements of the NF-κB pathway. CONCLUSIONS AND IMPLICATIONS We showed for the first time that hypoxia leads to a pathogenetic ligand-independent activation of hepatic MRs during cirrhosis resulting in their nuclear translocation and transcriptional activation of the NF-κB pathway. Treatment with eplerenone prevented the worsening of cirrhosis by blocking this ligand-independent activation of the MR.
Collapse
Affiliation(s)
- Barbara Schreier
- Julius Bernstein Institute of Physiology, Medical School, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Anja Wolf
- Laboratory of Molecular Hepatology, Clinic of Internal Medicine I, Martin Luther University of Halle-Wittenberg, Halle, Germany.,Julius Bernstein Institute of Physiology, Medical School, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Stefanie Hammer
- Laboratory of Molecular Hepatology, Clinic of Internal Medicine I, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Sabine Pohl
- Laboratory of Molecular Hepatology, Clinic of Internal Medicine I, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Sigrid Mildenberger
- Julius Bernstein Institute of Physiology, Medical School, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Sindy Rabe
- Julius Bernstein Institute of Physiology, Medical School, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Michael Gekle
- Julius Bernstein Institute of Physiology, Medical School, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Alexander Zipprich
- Laboratory of Molecular Hepatology, Clinic of Internal Medicine I, Martin Luther University of Halle-Wittenberg, Halle, Germany
| |
Collapse
|
13
|
Yavas G, Celik E, Yavas C, Elsurer C, Afsar RE. Spironolactone ameliorates the cardiovascular toxicity induced by concomitant trastuzumab and thoracic radiotherapy. Rep Pract Oncol Radiother 2017; 22:295-302. [PMID: 28507459 DOI: 10.1016/j.rpor.2017.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/28/2017] [Indexed: 02/08/2023] Open
Abstract
AIM We aimed to evaluate impact of spironolactone (S) on cardiovascular toxicity of concomitant use of radiotherapy (RT) and trastuzumab (T). BACKGROUND S, an aldosterone receptor antagonist, is known to ameliorate the cardiac damage. S ameliorates anthracycline -induced cardiotoxicity, there is no data regarding to effect of S on both T and radiation-induced cardiotoxicity. MATERIALS/METHODS Eighty rats were divided into eight groups: group (G) 1 was defined as control group. G2, G3 and G4 were RT, S and T groups respectively. G5, G6, G7 and G8 were RT + T, T + S, RT + S and RT + T + S groups respectively. Rats were sacrificed at 6th hour; 21st and 100th days after RT. Heart and thoracic aorta samples were taken for microscopical examination. RESULTS Cardiac inflammation and fibrosis scores and; TGF-β expression were not significantly different within study groups at 6th hour and 21st days of RT. By 100th days of RT fibrosis scores and TGF-β expression in cardiac samples were significantly different between study groups (p values were 0.004 and 0.002 respectively). Pair-wise comparisons revealed that both cardiac fibrosis scores and TGF-β expression levels were higher in G5 when compared to G8 (p values were 0.046 and 0.028 respectively). Moreover the TGF-β expression was higher in G5 when compared to G2 (p = 0.046). We could not demonstrate any significant differences with respect to inflammation, fibrosis and TGF-β expression in thoracic aorta samples between study groups. CONCLUSIONS Although S had a protective effect on cardiac tissue it had no protective effect on thoracic aorta when administered with RT + T.
Collapse
Affiliation(s)
- Guler Yavas
- Selcuk University, Department of Radiation Oncology, Konya, Turkey
| | - Esin Celik
- Selcuk University, Department of Pathology, Konya, Turkey
| | - Cagdas Yavas
- Selcuk University, Department of Radiation Oncology, Konya, Turkey
| | - Cagdas Elsurer
- Selcuk University, Department of Ear, Nose and Throat, Konya, Turkey
| | | |
Collapse
|
14
|
Ong GSY, Young MJ. Mineralocorticoid regulation of cell function: the role of rapid signalling and gene transcription pathways. J Mol Endocrinol 2017; 58:R33-R57. [PMID: 27821439 DOI: 10.1530/jme-15-0318] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/06/2016] [Indexed: 12/22/2022]
Abstract
The mineralocorticoid receptor (MR) and mineralocorticoids regulate epithelial handling of electrolytes, and induces diverse effects on other tissues. Traditionally, the effects of MR were ascribed to ligand-receptor binding and activation of gene transcription. However, the MR also utilises a number of intracellular signalling cascades, often by transactivating unrelated receptors, to change cell function more rapidly. Although aldosterone is the physiological mineralocorticoid, it is not the sole ligand for MR. Tissue-selective and mineralocorticoid-specific effects are conferred through the enzyme 11β-hydroxysteroid dehydrogenase 2, cellular redox status and properties of the MR itself. Furthermore, not all aldosterone effects are mediated via MR, with implication of the involvement of other membrane-bound receptors such as GPER. This review will describe the ligands, receptors and intracellular mechanisms available for mineralocorticoid hormone and receptor signalling and illustrate their complex interactions in physiology and disease.
Collapse
Affiliation(s)
- Gregory S Y Ong
- Cardiovascular Endocrinology LaboratoryCentre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of MedicineSchool of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Morag J Young
- Cardiovascular Endocrinology LaboratoryCentre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of PhysiologySchool of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
15
|
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
|
16
|
Shieh FK, Kotlyar E, Sam F. Aldosterone and cardiovascular remodelling: focus on myocardial failure. J Renin Angiotensin Aldosterone Syst 2016; 5:3-13. [PMID: 15136967 DOI: 10.3317/jraas.2004.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Heart failure is a clinical syndrome that may result from different disease states or conditions that injure the myocardium. The activation of circulating neurohormones, particularly aldosterone, may play a pivotal role in left ventricular (LV) remodelling. The Randomized Aldactone Evaluation Study and Eplerenone Post-Acute Myocardial Infarction Heart Failure Efficacy and Survival trial have emphasised the clinical importance of aldosterone. This review addresses some of the proposed mechanisms of LV remodelling in heart failure.
Collapse
Affiliation(s)
- Frederick K Shieh
- Boston University Medical School, Boston University Medical Center, Boston, Massechussetts, USA
| | | | | |
Collapse
|
17
|
D'Uva G, Lauriola M. Towards the emerging crosstalk: ERBB family and steroid hormones. Semin Cell Dev Biol 2015; 50:143-52. [PMID: 26582250 DOI: 10.1016/j.semcdb.2015.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 10/28/2015] [Accepted: 11/09/2015] [Indexed: 01/05/2023]
Abstract
Growth factors acting through receptor tyrosine kinases (RTKs) of ERBB family, along with steroid hormones (SH) acting through nuclear receptors (NRs), are critical signalling mediators of cellular processes. Deregulations of ERBB and steroid hormone receptors are responsible for several diseases, including cancer, thus demonstrating the central role played by both systems. This review will summarize and shed light on an emerging crosstalk between these two important receptor families. How this mutual crosstalk is attained, such as through extensive genomic and non-genomic interactions, will be addressed. In light of recent studies, we will describe how steroid hormones are able to fine-tune ERBB feedback loops, thus impacting on cellular output and providing a new key for understanding the complexity of biological processes in physiological or pathological conditions. In our understanding, the interactions between steroid hormones and RTKs deserve further attention. A system biology approach and advanced technologies for the analysis of RTK-SH crosstalk could lead to major advancements in molecular medicine, providing the basis for new routes of pharmacological intervention in several diseases, including cancer.
Collapse
Affiliation(s)
- Gabriele D'Uva
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Mattia Lauriola
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Experimental, Diagnostic and Specialty Medicine - DIMES, University of Bologna, Bologna 40138, Italy.
| |
Collapse
|
18
|
Miliukov VE, Bogdanov AV, Murshudova KM, Nguen KK, Polunin SV, Zharikov IO. [Role of morphofunctional adrenal changes in pathogenesis of water-electrolyte disorders in patients with acute intestinal obstruction]. Khirurgiia (Mosk) 2015:90-95. [PMID: 25942764 DOI: 10.17116/hirurgia2015190-95] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- V E Miliukov
- Kafedra anatomii cheloveka Pervogo Moskovskogo gosudarstvennogo meditsinskogo universiteta im. I.M. Sechenova Minzdrava RF; Kafedra voenno-polevoĭ khirurgii Instituta usovershenstvovaniia vracheĭ Meditsinskogo uchebno-nauchnogo klinicheskogo tsentra im. P.V. Mandryka MO RF
| | - A V Bogdanov
- Kafedra voenno-polevoĭ khirurgii Instituta usovershenstvovaniia vracheĭ Meditsinskogo uchebno-nauchnogo klinicheskogo tsentra im. P.V. Mandryka MO RF
| | - Kh M Murshudova
- Kafedra anatomii cheloveka Pervogo Moskovskogo gosudarstvennogo meditsinskogo universiteta im. I.M. Sechenova Minzdrava RF; Nauchno-issledovatel'skiĭ institut morfologii cheloveka RAMN
| | - K K Nguen
- Kafedra anatomii cheloveka Pervogo Moskovskogo gosudarstvennogo meditsinskogo universiteta im. I.M. Sechenova Minzdrava RF
| | - S V Polunin
- Kafedra voenno-polevoĭ khirurgii Instituta usovershenstvovaniia vracheĭ Meditsinskogo uchebno-nauchnogo klinicheskogo tsentra im. P.V. Mandryka MO RF
| | - Iu O Zharikov
- Nauchno-issledovatel'skiĭ institut skoroĭ pomoshchi im. N.V. Sklifosovskogo, Moskva
| |
Collapse
|
19
|
Response to ‘Oxidative stress in patients affected by primary aldosteronism’. J Hypertens 2015; 33:884. [DOI: 10.1097/hjh.0000000000000524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
|
21
|
Ferreira NS, Cau SBA, Silva MAB, Manzato CP, Mestriner FLAC, Matsumoto T, Carneiro FS, Tostes RC. Diabetes impairs the vascular effects of aldosterone mediated by G protein-coupled estrogen receptor activation. Front Pharmacol 2015; 6:34. [PMID: 25784875 PMCID: PMC4345803 DOI: 10.3389/fphar.2015.00034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/10/2015] [Indexed: 01/12/2023] Open
Abstract
Aldosterone promotes non-genomic effects in endothelial and vascular smooth muscle cells via activation of mineralocorticoid receptors (MR) and G protein-coupled estrogen receptors (GPER). GPER activation is associated with beneficial/protective effects in the vasculature. Considering that vascular dysfunction plays a major role in diabetes-associated complications, we hypothesized that the beneficial effects mediated by vascular GPER activation, in response to aldosterone, are decreased in diabetes. Mesenteric resistance arteries from female, 14-16 weeks-old, control and diabetic (db/db) mice were used. Phenylephrine (PhE)-induced contractions were greater in arteries from db/db vs. control mice. Aldosterone (10 nM) increased maximal contractile responses to PhE in arteries from control mice, an effect elicited via activation of GPER. Although aldosterone did not increase PhE responses in arteries from db/db mice, blockade of GPER, and MR decreased PhE-induced contractile responses in db/db mesenteric arteries. Aldosterone also reduced the potency of acetylcholine (ACh)-induced relaxation in arteries from both control and db/db mice via MR-dependent mechanisms. GPER antagonism further decreased ACh-induced relaxation in the control group, but did not affect ACh responses in the diabetic group. Aldosterone increased extracellular signal-regulated kinase 1/2 phosphorylation in arteries from control and db/db mice by a GPER-dependent mechanism. GPER, but not MR, gene, and protein expression, determined by RT-PCR and immunoblotting/immunofluorescence assays, respectively, were increased in arteries from db/db mice vs. control arteries. These findings indicate that aldosterone activates both vascular MR and GPER and that the beneficial effects of GPER activation are decreased in arteries from diabetic animals. Our results further elucidate the mechanisms by which aldosterone influences vascular function and contributes to vascular dysfunction in diabetes. Financial Support: FAPESP, CNPq, and CAPES, Brazil.
Collapse
Affiliation(s)
- Nathanne S Ferreira
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo São Paulo Brazil
| | - Stêfany B A Cau
- Department of Pharmacology, Biological Sciences Institute, Federal University of Minas Gerais Belo Horizonte, Brazil
| | - Marcondes A B Silva
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo São Paulo Brazil
| | - Carla P Manzato
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo São Paulo Brazil
| | - Fabíola L A C Mestriner
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo São Paulo Brazil
| | - Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University Tokyo, Japan
| | - Fernando S Carneiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo São Paulo Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo São Paulo Brazil
| |
Collapse
|
22
|
Meinel S, Gekle M, Grossmann C. Mineralocorticoid receptor signaling: crosstalk with membrane receptors and other modulators. Steroids 2014; 91:3-10. [PMID: 24928729 DOI: 10.1016/j.steroids.2014.05.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 05/16/2014] [Accepted: 05/28/2014] [Indexed: 12/30/2022]
Abstract
The mineralocorticoid receptor (MR) belongs to the steroid receptor superfamily. Classically, it acts as a ligand-bound transcription factor in epithelial tissues, where it regulates water and electrolyte homeostasis and controls blood pressure. Additionally, the MR has been shown to elicit pathophysiological effects including inflammation, fibrosis and remodeling processes in the cardiovascular system and the kidneys and MR antagonists have proven beneficial for patients with certain cardiovascular and renal disease. The underlying molecular mechanisms that mediate MR effects have not been fully elucidated but very likely rely on interactions with other signaling pathways in addition to genomic actions at hormone response elements. In this review we will focus on interactions of MR signaling with different membrane receptors, namely receptor tyrosine kinases and the angiotensin II receptor because of their potential relevance for disease. In addition, GPR30 is discussed as a new aldosterone receptor. To gain insights into the problem why the MR only seems to mediate pathophysiological effects in the presence of additional permissive factors we will also briefly discuss factors that lead to modulation of MR activity as well. Overall, MR signaling is part of an intricate network that still needs to be investigated further.
Collapse
Affiliation(s)
- S Meinel
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany
| | - M Gekle
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany
| | - C Grossmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany.
| |
Collapse
|
23
|
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
|
24
|
Abstract
Aldosterone regulates blood pressure through its effects on the kidney and the cardiovascular system. Dysregulation of aldosterone signalling can result in hypertension which in turn can lead to chronic pathologies of the kidney such as renal fibrosis and nephropathy. Aldosterone acts by binding to the mineralocorticoid receptor (MR), which acts as a ligand-dependent transcription factor in target tissues such as segments of the distal nephron including the connecting tubule and cortical collecting duct (CCD). Aldosterone also promotes the activation of protein kinase signalling cascades that are coupled to growth factor receptors and act directly on specific substrates in the cell membrane or cytoplasm. The rapid actions of aldosterone can also modulate gene expression through the phosphorylation of transcription factors. Aldosterone is a key regulator of Na(+) conservation in the distal nephron, largely through multiple mechanisms that modulate the activity of the epithelial Na(+) channel (ENaC). Aldosterone transcriptionally up-regulates the ENaCα subunit and also up regulates serum and glucocorticoid-regulated kinase-1 (SGK1) that indirectly regulates the ubiquitination of ENaC subunits. Aldosterone promotes the activation of protein kinase D1 (PKD1) which can modify the activity of ENaC and other transporters through effects on sub-cellular trafficking. In M1-CCD cells, early sub-cellular trafficking causes the redistribution of ENaC subunits within minutes of treatment with aldosterone. ENaC subunits can also interact directly with phosphatidylinositide signalling intermediates in the membrane and the mechanism by which PKD isoforms regulate protein trafficking is through the control of vesicle fission from the trans Golgi network by activation of phosphatidylinositol 4-kinaseIIIβ (PI4KIIIβ).
Collapse
Affiliation(s)
- Sinéad Quinn
- Molecular Medicine Laboratories, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Brian J Harvey
- Molecular Medicine Laboratories, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Warren Thomas
- Molecular Medicine Laboratories, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland.
| |
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
|
Atorvastatin Suppresses Aldosterone-induced Neonatal Rat Cardiac Fibroblast Proliferation by Inhibiting ERK1/2 in the Genomic Pathway. J Cardiovasc Pharmacol 2013; 61:520-7. [DOI: 10.1097/fjc.0b013e31828c090e] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Yavas G, Elsurer R, Yavas C, Elsurer C, Ata O. Does spironolactone ameliorate trastuzumab-induced cardiac toxicity? Med Hypotheses 2013; 81:231-4. [PMID: 23688742 DOI: 10.1016/j.mehy.2013.04.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 04/26/2013] [Indexed: 01/01/2023]
Abstract
The ErbB2 receptor is a proto-oncogene associated with a poor prognosis in breast cancer. Trastuzumab, a humanized anti-ErbB2 antibody currently in clinical use, has proven to be an essential tool in the immunotherapy of breast carcinoma. Additionally, ErbB2 is involved in the growth and survival pathway of adult cardiomyocytes which accounts for the trastuzumab-induced cardiotoxicity. Moreover, in metastatic breast cancer patients treated with trastuzumab, endomyocardial biopsy documented focal vacuolar changes, pleomorphic mitochondria, myocardial cell hypertrophy, and mild interstitial fibrosis on electron microscopy without accompanying light microscopic abnormalities, a finding consistent with a reversible pattern of cardiac injury. On the other hand, aldosterone and mineralocorticoid receptor (MR) researches have experienced a revival after the discovery that aldosterone and MR are not only involved in the electrolyte and volume balance but also in the pathophysiological processes of the reno-cardiovascular system. Aldosterone has both genomic and nongenotropic effects on epidermal growth factor receptor (EGFR) expression. Genomic effect induces genomic up-regulation of the EGFR protein expression via EGFR promoter, whereas nongenotropic effect leads to the EGFR transactivation resulting in persistent pathophysiological effects including formation of extracellular matrix and myocardial hypertrophy. Spironolactone, an aldosterone receptor antagonist, is known to ameliorate the cardiac damage. The underlying mechanism for the genomic interactions seem to be the stimulation of the EGFR promoter by aldosterone-bound MR, which then dose-dependently enhances the EGFR protein levels, which may be successively inhibited by spironolactone. By the light of these findings, we hypothesize that spironolactone may ameliorate trastuzumab-induced cardiotoxicity via inhibition of transactivation of the EGFR by aldosterone and reversing myocardial hypertrophy. This issue warrants further studies.
Collapse
Affiliation(s)
- Guler Yavas
- Department of Radiation Oncology, Selcuk University, Konya, Turkey.
| | | | | | | | | |
Collapse
|
28
|
Tiberio L, Nascimbeni R, Villanacci V, Casella C, Fra A, Vezzoli V, Furlan L, Meyer G, Parrinello G, Baroni MD, Salerni B, Schiaffonati L. The decrease of mineralcorticoid receptor drives angiogenic pathways in colorectal cancer. PLoS One 2013; 8:e59410. [PMID: 23555666 PMCID: PMC3610652 DOI: 10.1371/journal.pone.0059410] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 02/13/2013] [Indexed: 12/03/2022] Open
Abstract
Angiogenesis plays a crucial role in tumor growth and progression. Low expression of mineralocorticoid receptor (MR) in several malignant tumors correlates with disease recurrence and overall survival. Previous studies have shown that MR expression is decreased in colorectal cancer (CRC). Here we hypothesize that decreased MR expression can contribute to angiogenesis and poor patient survival in colorectal malignancies. In a cohort of CRC patients, we analyzed tumor MR expression, its correlation with tumor microvascular density and its impact on survival. Subsequently, we interrogated the role of MR in angiogenesis in an in vitro model, based on the colon cancer cell line HCT116, ingenierized to re-express a physiologically controlled MR. In CRC, decreased MR expression was associated with increased microvascular density and poor patient survival. In pchMR transfected HCT116, aldosterone or natural serum steroids largely inhibited mRNA expression levels of both VEGFA and its receptor 2/KDR. In CRC, MR activation may significantly decrease angiogenesis by directly inhibiting dysregulated VEGFA and hypoxia-induced VEGFA mRNA expression. In addition, MR activation attenuates the expression of the VEGF receptor 2/KDR, possibly dampening the activation of a VEGFA/KDR dependent signaling pathway important for the survival of tumor cells under hypoxic conditions.
Collapse
Affiliation(s)
- Laura Tiberio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- * E-mail: (MDB); (LT)
| | - Riccardo Nascimbeni
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- First Unit of General Surgery, Brescia City Hospital, Brescia, Italy
| | | | - Claudio Casella
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- First Unit of General Surgery, Brescia City Hospital, Brescia, Italy
| | - Anna Fra
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Valeria Vezzoli
- Department of BioSciences, University of Milano, Milan, Italy
| | - Lucia Furlan
- Department of Clinical and Experimental Medicine, University of Padova, Padova, Italy
| | - Giuliano Meyer
- Department of BioSciences, University of Milano, Milan, Italy
| | - Giovanni Parrinello
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Maurizio D. Baroni
- Department of Biology, University of Padova, Padova, Italy
- * E-mail: (MDB); (LT)
| | - Bruno Salerni
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- First Unit of General Surgery, Brescia City Hospital, Brescia, Italy
| | - Luisa Schiaffonati
- Department of Clinical and Experimental Medicine, University of Padova, Padova, Italy
| |
Collapse
|
29
|
Combined Effects of PPAR γ Agonists and Epidermal Growth Factor Receptor Inhibitors in Human Proximal Tubule Cells. PPAR Res 2013; 2013:982462. [PMID: 23533381 PMCID: PMC3596915 DOI: 10.1155/2013/982462] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 01/25/2013] [Indexed: 12/28/2022] Open
Abstract
We aimed to determine whether epidermal growth factor receptor (EGFR) inhibition, in addition to a peroxisome proliferator-activated receptor gamma (PPARγ) agonist, prevents high-glucose-induced proximal tubular fibrosis, inflammation, and sodium and water retention in human proximal tubule cells exposed to normal glucose; high glucose; high glucose with the PPARγ agonist pioglitazone or with the P-EGFR inhibitor, gefitinib; or high glucose with both pioglitazone and gefitinib. We have shown that high glucose increases AP-1 and NFκB binding activity, downstream phosphorylation of EGFR and Erk1/2, and fibronectin and collagen IV expression. Pioglitazone reversed these effects but upregulated NHE3 and AQP1 expression. Gefitinib inhibited high glucose induced fibronectin and collagen IV, and EGFR and Erk1/2 phosphorylation and reversed pioglitazone-induced increases in NHE3 and AQP1 expression. Our data suggests that combination of an EGFR inhibitor and a PPARγ agonist mitigates high-glucose-induced fibrosis and inflammation and reverses the upregulation of transporters and channels involved in sodium and water retention in human proximal tubule cells. Hence EGFR blockade may hold promise, not only in limiting tubulointerstitial pathology in diabetic nephropathy, but also in limiting the sodium and water retention observed in patients with diabetes and exacerbated by PPARγ agonists.
Collapse
|
30
|
Ruhs S, Strätz N, Schlör K, Meinel S, Mildenberger S, Rabe S, Gekle M, Grossmann C. Modulation of transcriptional mineralocorticoid receptor activity by nitrosative stress. Free Radic Biol Med 2012; 53:1088-100. [PMID: 22749806 DOI: 10.1016/j.freeradbiomed.2012.06.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 05/29/2012] [Accepted: 06/19/2012] [Indexed: 11/18/2022]
Abstract
The mineralocorticoid receptor (MR) plays an important role in salt and water homeostasis and pathological tissue modifications, such as cardiovascular and renal fibrosis. Importantly, MR activation by aldosterone per se is not sufficient for the deleterious effects but requires the additional presence of a certain pathological milieu. Phenomenologically, this milieu could be generated by enhanced nitrosative stress. However, little is known regarding the modulation of MR transcriptional activity in a pathological milieu. The glucocorticoid receptor (GR), the closest relative of the MR, binds to the same hormone-response element but elicits protective effects on the cardiovascular system. To investigate the possible modulation of MR and GR by nitrosative stress under controlled conditions we used human embryonic kidney (HEK) cells and measured MR and GR transactivation after stimulation with the nitric oxide (NO)-donor SNAP and the peroxynitrite-donor Sin-1. In the presence of corticosteroids NO led to a general reduced corticosteroid receptor activity by repression of corticosteroid receptor-DNA interaction. The NO-induced diminished transcriptional MR activity was most pronounced during stimulation with physiological aldosterone concentrations, suggesting that NO treatment prevented its pathophysiological overactivation. In contrast, single peroxynitrite administration specifically induced the MR transactivation activity whereas genomic GR activity remained unchanged. Mechanistically, peroxynitrite permitted nuclear MR translocation whereas the cytosolic GR distribution was unaffected. Consequently, peroxynitrite represents a MR-specific aldosterone mimetic. In summary, our data indicate that the genomic function of corticosteroid receptors can be modulated by nitrosative stress which may induce the shift from physiological toward pathophysiological MR effects.
Collapse
Affiliation(s)
- Stefanie Ruhs
- Julius-Bernstein-Institut für Physiologie der Universität Halle-Wittenberg, Halle, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Faresse N, Vitagliano JJ, Staub O. Differential ubiquitylation of the mineralocorticoid receptor is regulated by phosphorylation. FASEB J 2012; 26:4373-82. [PMID: 22798426 DOI: 10.1096/fj.12-209924] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Aldosterone stimulation of the mineralocorticoid receptor (MR) is involved in numerous physiological responses, including Na+ homeostasis, blood pressure control, and heart failure. Aldosterone binding to MR promotes different post-translational modifications that regulate MR nuclear translocation, gene expression, and finally receptor degradation. Here, we show that aldosterone stimulates rapid phosphorylation of MR via ERK1/2 in a dose-dependent manner (from 0.1 to 10 nM) in renal epithelial cells. This phosphorylation induces an increase of MR apparent molecular weight, with a maximal upward shift of 30 kDa. Strikingly, these modifications are critical for the regulation of the MR ubiquitylation state. Indeed, we find that MR is monoubiquitylated in its basal state, and this status is sustained by the tumor suppressor gene 101 (Tsg101). Phosphorylation leads to disruption of MR/Tsg101 association and monoubiquitin removal. These events prompt polyubiquitin-dependent destabilization of MR and degradation. Preventing MR phosphorylation by ERK1/2 inhibition or mutation of target serines affects the sequential mechanisms of MR ubiquitylation and inhibits the aldosterone-mediated degradation. Our data provide a novel model of negative feedback of aldosterone signaling, involving sequential phosphorylation, monoubiquitin removal and subsequent polyubiquitylation/degradation of MR.
Collapse
Affiliation(s)
- Nourdine Faresse
- University of Lausanne, Department of Pharmacology and Toxicology, Rue du Bugnon 27 CH-1005 Lausanne, Switzerland
| | | | | |
Collapse
|
32
|
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
|
33
|
Schwerdt G, Frisch A, Mildenberger S, Hilgenfeld T, Grossmann C, Gekle M. Influence of aldosterone and salt or ouabain in a10 rat aorta smooth muscle cells. J Vasc Res 2012; 49:231-41. [PMID: 22433677 DOI: 10.1159/000334091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 09/28/2011] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND/AIMS It is currently under debate whether aldosterone is able to induce fibrosis or whether it acts only as a cofactor under pathological conditions, e.g. as an elevated salt (NaCl) load. METHODS We tested the interaction of 10 nM aldosterone, 15 mM NaCl and 1 μM ouabain using rat aorta smooth muscle cells (A10) with respect to the following parameters: necrosis, apoptosis, glucose-6-phosphate dehydrogenase (G6PD) and 6-phosphogluconate dehydrogenase activity, glutathione (GSH) content, collagen and fibronectin homeostasis and intracellular calcium distribution. RESULTS Necrosis rates were increased after 48 h of incubation with aldosterone, salt or ouabain and in the combination of aldosterone and salt or ouabain. Apoptosis rates were decreased. A reduced defense capacity against oxidative stress was mirrored in the decreased G6PD activity and GSH content. Collagen III or fibronectin synthesis rates were unchanged, but gelatinase activity was increased resulting in a decreased media collagen III and fibronectin content. Calcium stores were increased by aldosterone in combination with ouabain. CONCLUSION Aldosterone and salt per se can lead to cell injury that is aggravated in combination or with cardiotonic steroids. In cooperation with other vascular cells, this can generate a permissive milieu enabling aldosterone or salt to promote more extensive vascular injury.
Collapse
Affiliation(s)
- Gerald Schwerdt
- Julius-Bernstein-Institut für Physiologie, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany.
| | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Seiferth A, Ruhs S, Mildenberger S, Gekle M, Grossmann C. The phosphatase calcineurin PP2BAβ mediates part of mineralocorticoid receptor transcriptional activity. FASEB J 2012; 26:2327-37. [DOI: 10.1096/fj.11-199976] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Anja Seiferth
- Julius‐Bernstein‐Institut für PhysiologieUniversität Halle‐WittenbergHalleGermany
| | - Stefanie Ruhs
- Julius‐Bernstein‐Institut für PhysiologieUniversität Halle‐WittenbergHalleGermany
| | - Sigrid Mildenberger
- Julius‐Bernstein‐Institut für PhysiologieUniversität Halle‐WittenbergHalleGermany
| | - Michael Gekle
- Julius‐Bernstein‐Institut für PhysiologieUniversität Halle‐WittenbergHalleGermany
| | - Claudia Grossmann
- Julius‐Bernstein‐Institut für PhysiologieUniversität Halle‐WittenbergHalleGermany
| |
Collapse
|
36
|
Messaoudi S, Zhang AD, Griol-Charhbili V, Escoubet B, Sadoshima J, Farman N, Jaisser F. The epidermal growth factor receptor is involved in angiotensin II but not aldosterone/salt-induced cardiac remodelling. PLoS One 2012; 7:e30156. [PMID: 22291909 PMCID: PMC3264592 DOI: 10.1371/journal.pone.0030156] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 12/11/2011] [Indexed: 01/10/2023] Open
Abstract
Experimental and clinical studies have shown that aldosterone/mineralocorticoid receptor (MR) activation has deleterious effects in the cardiovascular system; however, the signalling pathways involved in the pathophysiological effects of aldosterone/MR in vivo are not fully understood. Several in vitro studies suggest that Epidermal Growth Factor Receptor (EGFR) plays a role in the cardiovascular effects of aldosterone. This hypothesis remains to be demonstrated in vivo. To investigate this question, we analyzed the molecular and functional consequences of aldosterone exposure in a transgenic mouse model with constitutive cardiomyocyte-specific overexpression of a mutant EGFR acting as a dominant negative protein (DN-EGFR). As previously reported, Angiotensin II-mediated cardiac remodelling was prevented in DN-EGFR mice. However, when chronic MR activation was induced by aldosterone-salt-uninephrectomy, cardiac hypertrophy was similar between control littermates and DN-EGFR. In the same way, mRNA expression of markers of cardiac remodelling such as ANF, BNF or β-Myosin Heavy Chain as well as Collagen 1a and 3a was similarly induced in DN-EGFR mice and their CT littermates. Our findings confirm the role of EGFR in AngII mediated cardiac hypertrophy, and highlight that EGFR is not involved in vivo in the damaging effects of aldosterone on cardiac function and remodelling.
Collapse
Affiliation(s)
- Smail Messaoudi
- INSERM, U872, Centre de Recherche des Cordeliers, Paris, France
- Pierre et Marie Curie University, Paris VI, Paris, France
| | - An Di Zhang
- INSERM, U872, Centre de Recherche des Cordeliers, Paris, France
- Pierre et Marie Curie University, Paris VI, Paris, France
| | - Violaine Griol-Charhbili
- INSERM, U872, Centre de Recherche des Cordeliers, Paris, France
- Pierre et Marie Curie University, Paris VI, Paris, France
| | - Brigitte Escoubet
- INSERM, U872, Centre de Recherche des Cordeliers, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Paris, France
- University Denis Diderot, Paris 7, Paris, France
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, New Jersey, United States of America
| | - Nicolette Farman
- INSERM, U872, Centre de Recherche des Cordeliers, Paris, France
- Pierre et Marie Curie University, Paris VI, Paris, France
| | - Frederic Jaisser
- INSERM, U872, Centre de Recherche des Cordeliers, Paris, France
- Pierre et Marie Curie University, Paris VI, Paris, France
- * E-mail:
| |
Collapse
|
37
|
Winter C, Kampik NB, Vedovelli L, Rothenberger F, Paunescu TG, Stehberger PA, Brown D, John H, Wagner CA. Aldosterone stimulates vacuolar H(+)-ATPase activity in renal acid-secretory intercalated cells mainly via a protein kinase C-dependent pathway. Am J Physiol Cell Physiol 2011; 301:C1251-61. [PMID: 21832245 DOI: 10.1152/ajpcell.00076.2011] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Urinary acidification in the collecting duct is mediated by the activity of H(+)-ATPases and is stimulated by various factors including angiotensin II and aldosterone. Classically, aldosterone effects are mediated via the mineralocorticoid receptor. Recently, we demonstrated a nongenomic stimulatory effect of aldosterone on H(+)-ATPase activity in acid-secretory intercalated cells of isolated mouse outer medullary collecting ducts (OMCD). Here we investigated the intracellular signaling cascade mediating this stimulatory effect. Aldosterone stimulated H(+)-ATPase activity in isolated mouse and human OMCDs. This effect was blocked by suramin, a general G protein inhibitor, and GP-2A, a specific G(αq) inhibitor, whereas pertussis toxin was without effect. Inhibition of phospholipase C with U-73122, chelation of intracellular Ca(2+) with BAPTA, and blockade of protein kinase C prevented the stimulation of H(+)-ATPases. Stimulation of PKC by DOG mimicked the effect of aldosterone on H(+)-ATPase activity. Similarly, aldosterone and DOG induced a rapid translocation of H(+)-ATPases to the luminal side of OMCD cells in vivo. In addition, PD098059, an inhibitor of ERK1/2 activation, blocked the aldosterone and DOG effects. Inhibition of PKA with H89 or KT2750 prevented and incubation with 8-bromoadenosine-cAMP mildly increased H(+)-ATPase activity. Thus, the nongenomic modulation of H(+)-ATPase activity in OMCD-intercalated cells by aldosterone involves several intracellular pathways and may be mediated by a G(αq) protein-coupled receptor and PKC. PKA and cAMP appear to have a modulatory effect. The rapid nongenomic action of aldosterone may participate in the regulation of H(+)-ATPase activity and contribute to final urinary acidification.
Collapse
Affiliation(s)
- Christian Winter
- Institute of Physiology, Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Panchapakesan U, Pollock C, Saad S. Renal epidermal growth factor receptor: its role in sodium and water homeostasis in diabetic nephropathy. Clin Exp Pharmacol Physiol 2011; 38:84-8. [PMID: 21155863 DOI: 10.1111/j.1440-1681.2010.05472.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
1. Volume expansion is observed in animal and human models of diabetic nephropathy, which is in a large part a result of disordered renal tubular cell sodium and water transport. 2. Sodium transport in the proximal tubule is increased in diabetes mellitus as a result of enhanced activity of the sodium-hydrogen exchanger-3 (NHE3), the key transporter for transcellular reabsorption of sodium. Transactivation of the epidermal growth factor receptor (EGFR) by factors inherent in the milieu of diabetes mellitus increases serum glucocorticoid regulated kinase-1 (Sgk1), a key regulator of NHE3. 3. Enhanced sodium and water reabsorption, occurring as a consequence of endogenous or pharmacological stimulation of the peroxisome proliferator-activated receptor gamma is Sgk1 mediated. 4. EGFR inhibitors, which are currently used clinically to treat malignancies, might have potential in attenuating the cellular mechanisms responsible for thiazolidinedione (TZD)-mediated sodium and water transport in diabetes. 5. In the present review, the authors focus on the importance of the EGFR in sodium and water uptake in the proximal tubule in the environment of pathophysiological and pharmacological influences.
Collapse
Affiliation(s)
- Usha Panchapakesan
- Renal Research Group, Department of Medicine, University of Sydney, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | | | | |
Collapse
|
39
|
Acidic environment leads to ROS-induced MAPK signaling in cancer cells. PLoS One 2011; 6:e22445. [PMID: 21818325 PMCID: PMC3144229 DOI: 10.1371/journal.pone.0022445] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2011] [Accepted: 06/21/2011] [Indexed: 02/07/2023] Open
Abstract
Tumor micromilieu often shows pronounced acidosis forcing cells to adapt their phenotype towards enhanced tumorigenesis induced by altered cellular signalling and transcriptional regulation. In the presents study mechanisms and potential consequences of the crosstalk between extra- and intracellular pH (pHe, pHi) and mitogen-activated-protein-kinases (ERK1/2, p38) was analyzed. Data were obtained mainly in AT1 R-3327 prostate carcinoma cells, but the principle importance was confirmed in 5 other cell types. Extracellular acidosis leads to a rapid and sustained decrease of pHi in parallel to p38 phosphorylation in all cell types and to ERK1/2 phosphorylation in 3 of 6 cell types. Furthermore, p38 phosphorylation was elicited by sole intracellular lactacidosis at normal pHe. Inhibition of ERK1/2 phosphorylation during acidosis led to necrotic cell death. No evidence for the involvement of the kinases c-SRC, PKC, PKA, PI3K or EGFR nor changes in cell volume in acidosis-induced MAPK activation was obtained. However, our data reveal that acidosis enhances the formation of reactive oxygen species (ROS), probably originating from mitochondria, which subsequently trigger MAPK phosphorylation. Scavenging of ROS prevented acidosis-induced MAPK phosphorylation whereas addition of H2O2 enhanced it. Finally, acidosis increased phosphorylation of the transcription factor CREB via p38, leading to increased transcriptional activity of a CRE-reporter even 24 h after switching the cells back to a normal environmental milieu. Thus, an acidic tumor microenvironment can induce a longer lasting p38-CREB-medited change in the transcriptional program, which may maintain the altered phenotype even when the cells leave the tumor environment.
Collapse
|
40
|
Abstract
The steroid hormone aldosterone is a key regulator of electrolyte transport in the kidney and contributes to both homeostatic whole-body electrolyte balance and the development of renal and cardiovascular pathologies. Aldosterone exerts its action principally through the mineralocorticoid receptor (MR), which acts as a ligand-dependent transcription factor in target tissues. Aldosterone also stimulates the activation of protein kinases and secondary messenger signaling cascades that act independently on specific molecular targets in the cell membrane and also modulate the transcriptional action of aldosterone through MR. This review describes current knowledge regarding the mechanisms and targets of rapid aldosterone action in the nephron and how aldosterone integrates these responses into the regulation of renal physiology.
Collapse
Affiliation(s)
- Warren Thomas
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland.
| | | |
Collapse
|
41
|
Callera GE, Yogi A, Briones AM, Montezano AC, He Y, Tostes RC, Schiffrin EL, Touyz RM. Vascular proinflammatory responses by aldosterone are mediated via c-Src trafficking to cholesterol-rich microdomains: role of PDGFR. Cardiovasc Res 2011; 91:720-31. [DOI: 10.1093/cvr/cvr131] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
42
|
Ziegler CG, Langbein H, Krug AW, Ludwig B, Eisenhofer G, Ehrhart-Bornstein M, Bornstein SR. Direct effect of dehydroepiandrosterone sulfate (DHEAS) on PC-12 cell differentiation processes. Mol Cell Endocrinol 2011; 336:149-55. [PMID: 21195131 DOI: 10.1016/j.mce.2010.12.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 12/03/2010] [Accepted: 12/21/2010] [Indexed: 11/24/2022]
Abstract
Dehydroepiandrosterone sulfate is classically seen as an inactive reservoir for the production of dehydroepiandrosterone. Steroid sulfatase is the enzyme that catalyzes the hydrolysis of dehydroepiandrosterone sulfate to dehydroepiandrosterone, which can then be further metabolized to other steroid hormones. Recent studies, however, indicate that dehydroepiandrosterone sulfate can mediate biological effects without being converted to dehydroepiandrosterone. This study aims to evaluate whether dehydroepiandrosterone sulfate itself influences the differentiation of PC-12 cells or if its desulfation to dehydroepiandrosterone is required. dehydroepiandrosterone and dehydroepiandrosterone sulfate both influence the differentiation of chromaffin PC-12 cells. Blocking steroid sulfatase activity and thereby the conversion of dehydroepiandrosterone sulfate to dehydroepiandrosterone by the enzyme blocker estrone sulfamate showed that the effect of dehydroepiandrosterone sulfate is independent of its conversion to dehydroepiandrosterone. Dehydroepiandrosterone sulfate, similar to dehydroepiandrosterone, reduced nerve growth factor-induced neurite outgrowth of PC-12 cells and the expression of synaptosomal-associated membrane protein of 25 kDa, increased the expression of chromogranin A and significantly increased dopamine release of PC-12 cells. In addition, dehydroepiandrosterone sulfate, dehydroepiandrosterone and membrane impermeable dehydroepiandrosterone-BSA all significantly reduced NGF-induced MAPK ERK1/2 signaling after 5 min. In summary, this study provides evidence that dehydroepiandrosterone sulfate, independent of its conversion to dehydroepiandrosterone, directs PC-12 cells' differentiation to a neuroendocrine direction. Furthermore, employing membrane-impermeable dehydroepiandrosterone-BSA indicates the involvement of plasma-membrane bound receptors.
Collapse
Affiliation(s)
- Christian G Ziegler
- Carl Gustav Carus University Hospital, Medical Clinic III, University of Dresden, Fetscherstr. 74, 01307 Dresden, Germany.
| | | | | | | | | | | | | |
Collapse
|
43
|
Griol-Charhbili V, Fassot C, Messaoudi S, Perret C, Agrapart V, Jaisser F. Epidermal Growth Factor Receptor Mediates the Vascular Dysfunction But Not the Remodeling Induced by Aldosterone/Salt. Hypertension 2011; 57:238-44. [DOI: 10.1161/hypertensionaha.110.153619] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pathophysiological aldosterone (aldo)/mineralocorticoid receptor signaling has a major impact on the cardiovascular system, resulting in hypertension and vascular remodeling. Mineralocorticoids induce endothelial dysfunction, decreasing vasorelaxation in response to acetylcholine and increasing the response to vasoconstrictors. Activation of the epidermal growth factor receptor (EGFR) is thought to mediate the vascular effects of aldo, but this has yet to be demonstrated in vivo. In this study, we analyzed the molecular and functional vascular consequences of aldo-salt challenge in the waved 2 mouse, a genetic model with a partial loss of EGFR tyrosine kinase activity. Deficient EGFR activity is associated with global oxidative stress and endothelial dysfunction. A decrease in EGFR activity did not affect the arterial wall remodeling process induced by aldo-salt. By contrast, normal EGFR activity was required for the aldo-induced enhancement of phenylephrine- and angiotensin II–mediated vasoconstriction. In conclusion, this in vivo study demonstrates that EGFR plays a key role in aldosterone-mediated vascular reactivity.
Collapse
Affiliation(s)
- Violaine Griol-Charhbili
- From the Institut National de la Santé et de la Recherche Médicale, U872, Centre de Recherche des Cordeliers, Paris, France; Pierre et Marie Curie University, Paris, France
| | - Céline Fassot
- From the Institut National de la Santé et de la Recherche Médicale, U872, Centre de Recherche des Cordeliers, Paris, France; Pierre et Marie Curie University, Paris, France
| | - Smail Messaoudi
- From the Institut National de la Santé et de la Recherche Médicale, U872, Centre de Recherche des Cordeliers, Paris, France; Pierre et Marie Curie University, Paris, France
| | - Claudine Perret
- From the Institut National de la Santé et de la Recherche Médicale, U872, Centre de Recherche des Cordeliers, Paris, France; Pierre et Marie Curie University, Paris, France
| | - Vincent Agrapart
- From the Institut National de la Santé et de la Recherche Médicale, U872, Centre de Recherche des Cordeliers, Paris, France; Pierre et Marie Curie University, Paris, France
| | - Frederic Jaisser
- From the Institut National de la Santé et de la Recherche Médicale, U872, Centre de Recherche des Cordeliers, Paris, France; Pierre et Marie Curie University, Paris, France
| |
Collapse
|
44
|
Leite-Dellova DCA, Malnic G, Mello-Aires M. Genomic and nongenomic stimulatory effect of aldosterone on H+-ATPase in proximal S3 segments. Am J Physiol Renal Physiol 2010; 300:F682-91. [PMID: 21190948 DOI: 10.1152/ajprenal.00172.2010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The genomic and nongenomic effects of aldosterone on the intracellular pH recovery rate (pHirr) via H(+)-ATPase and on cytosolic free calcium concentration ([Ca(2+)](i)) were investigated in isolated proximal S3 segments of rats during superfusion with an Na(+)-free solution, by using the fluorescent probes BCECF-AM and FLUO-4-AM, respectively. The pHirr, after cellular acidification with a NH(4)Cl pulse, was 0.064 ± 0.003 pH units/min (n = 17/74) and was abolished with concanamycin. Aldosterone (10(-12), 10(-10), 10(-8), or 10(-6) M with 1-h or 15- or 2-min preincubation) increased the pHirr. The baseline [Ca(2+)](i) was 103 ± 2 nM (n = 58). After 1 min of aldosterone preincubation, there was a transient and dose-dependent increase in [Ca(2+)](i) and after 6-min preincubation there was a new increase in [Ca(2+)](i) that persisted after 1 h. Spironolactone [mineralocorticoid (MR) antagonist], actinomycin D, or cycloheximide did not affect the effects of aldosterone (15- or 2-min preincubation) on pHirr and on [Ca(2+)](i) but inhibited the effects of aldosterone (1-h preincubation) on these parameters. RU 486 [glucocorticoid (GR) antagonist] and dimethyl-BAPTA (Ca(2+) chelator) prevented the effect of aldosterone on both parameters. The data indicate a genomic (1 h, via MR) and a nongenomic action (15 or 2 min, probably via GR) on the H(+)-ATPase and on [Ca(2+)](i). The results are compatible with stimulation of the H(+)-ATPase by increases in [Ca(2+)](i) (at 10(-12)-10(-6) M aldosterone) and inhibition of the H(+)-ATPase by decreases in [Ca(2+)](i) (at 10(-12) or 10(-6) M aldosterone plus RU 486).
Collapse
Affiliation(s)
- D C A Leite-Dellova
- Department of Basic Sciences, Faculdade de Zootecnia e Engenharia de Alimentos, Campus of Pirassununga, Instituto de Ciências Biomédicas, University of São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
45
|
Thum T, Schmitter K, Fleissner F, Wiebking V, Dietrich B, Widder JD, Jazbutyte V, Hahner S, Ertl G, Bauersachs J. Impairment of endothelial progenitor cell function and vascularization capacity by aldosterone in mice and humans. Eur Heart J 2010; 32:1275-86. [PMID: 20926363 PMCID: PMC3094546 DOI: 10.1093/eurheartj/ehq254] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Aims Hyperaldosteronism is associated with vascular injury and increased cardiovascular events. Bone marrow-derived endothelial progenitor cells (EPCs) play an important role in endothelial repair and vascular homeostasis. We hypothesized that hyperaldosteronism impairs EPC function and vascularization capacity in mice and humans. Methods and results We characterized the effects of aldosterone and mineralocorticoid receptor (MR) blockade on EPC number and function as well as vascularization capacity and endothelial function. Treatment of human EPC with aldosterone induced translocation of the MR and impaired multiple cellular functions of EPC, such as differentiation, migration, and proliferation in vitro. Impaired EPC function was rescued by pharmacological blockade or genetic ablation of the MR. Aldosterone protein kinase A (PKA) dependently increased reactive oxygen species formation in EPC. Aldosterone infusion in mice impaired EPC function, EPC homing to vascular structures and vascularization capacity in a MR-dependent but blood pressure-independent manner. Endothelial progenitor cells from patients with primary hyperaldosteronism compared with controls of similar age displayed reduced migratory potential. Impaired EPC function was associated with endothelial dysfunction. MR blockade in patients with hyperaldosteronism improved EPC function and arterial stiffness. Conclusion Endothelial progenitor cells express a MR that mediates functional impairment by PKA-dependent increase of reactive oxygen species. Normalization of EPC function may represent a novel mechanism contributing to the beneficial effects of MR blockade in cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Thomas Thum
- Institut für Molekulare und Translationale Therapiestrategien, Medizinische Hochschule Hannover, Germany
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Germany
- Corresponding author. Tel: +49 511 532 5272 (T.T), +49 511 532 3840 (J.B.); Fax: +49 511 532 5274 (T.T.), +49 511 532 5412 (J.B.), (T.T.), (J.B.)
| | - Kerstin Schmitter
- Institut für Molekulare und Translationale Therapiestrategien, Medizinische Hochschule Hannover, Germany
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
| | - Felix Fleissner
- Institut für Molekulare und Translationale Therapiestrategien, Medizinische Hochschule Hannover, Germany
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Germany
| | - Volker Wiebking
- Institut für Molekulare und Translationale Therapiestrategien, Medizinische Hochschule Hannover, Germany
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
| | - Bernd Dietrich
- Institut für Molekulare und Translationale Therapiestrategien, Medizinische Hochschule Hannover, Germany
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
| | - Julian D. Widder
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
| | - Virginija Jazbutyte
- Institut für Molekulare und Translationale Therapiestrategien, Medizinische Hochschule Hannover, Germany
| | - Stefanie Hahner
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
| | - Georg Ertl
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
| | - Johann Bauersachs
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Germany
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
- Corresponding author. Tel: +49 511 532 5272 (T.T), +49 511 532 3840 (J.B.); Fax: +49 511 532 5274 (T.T.), +49 511 532 5412 (J.B.), (T.T.), (J.B.)
| |
Collapse
|
46
|
Wei H, Mi X, Ji L, Yang L, Xia Q, Wei Y, Miyamori I, Fan C. Protein kinase C-delta is involved in induction of NOX1 gene expression by aldosterone in rat vascular smooth muscle cells. BIOCHEMISTRY (MOSCOW) 2010; 75:304-9. [PMID: 20370608 DOI: 10.1134/s0006297910030065] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In this study, we focused on the relationship between aldosterone and NOX1 expression in vascular smooth muscle cells (VSMCs). For the first time, with the use of specific inhibitors of protein kinase C (PKC), we report that PKCdelta mediates upregulation of NOX1 induced by 10 nM aldosterone in cultured VSMCs. Participation of PKC in the mediation of NOX1 regulation was further confirmed by the effect of diacylglycerol, a PKC agonist, on the NOX1 RNA in A7r5 cells with Northern blot analysis. To establish cause and effect, we next silenced the PKCdelta gene partly by RNA interference and found knockdown of PKCdelta gene attenuated aldosterone-induced NOX1 expression, generation of superoxide, as well as protein synthesis in VSMCs. Taken together, these data indicated PKCdelta might mediate aldosterone-dependent NOX1 upregulation in VSMCs. In addition, we showed that the cascade from aldosterone to PKCdelta activation had the participation of the mineralocorticoid receptor.
Collapse
Affiliation(s)
- Haiyan Wei
- State Key Laboratory of Biotherapy, West China Hospital and School of Life Science, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Limor R, Kaplan M, Sharon O, Knoll E, Naidich M, Weisinger G, Keidar S, Stern N. Aldosterone up-regulates 12- and 15-lipoxygenase expression and LDL oxidation in human vascular smooth muscle cells. J Cell Biochem 2010; 108:1203-10. [PMID: 19795383 DOI: 10.1002/jcb.22352] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Several lines of evidence suggest that aldosterone excess may have detrimental effects in the cardiovascular system, independent of its interaction with the renal epithelial cells. Here we examined the possibility that aldosterone modulates 12- and/or 15-lipoxygenase (LO) expression/activity in human vascular smooth muscle cells (VSMC), in vitro, thereby potentially contributing to both vascular reactivity and atherogenesis. Following 24 h treatment of VSMC with aldosterone (1 nmol/L), there was a approximately 2-fold increase in the generation rate of 12 hydroxyeicosatetraenoic acid (12-HETE), 70% increase in platelet type 12-LO mRNA expression (P < 0.001) along with a approximately 3-fold increase in 12-LO protein expression, which were blocked by the mineralocorticoid receptor (MR) antagonists spironolactone (100 nmol/L) and eplerelone (100 nmol/ml). Additionally, aldosterone (1 nmol/L; 24 h) increased the production of 15-HETE (50%; P < 0.001) and the expression of 15-LO type 2 mRNA (50%; P < 0.05) (in VSMC). Aldosterone also increased the 12- and 15-LO type 2 mRNA expression in a line of human aortic smooth muscle cells (T/G HA-VSMC) (60% and 50%, respectively). Aldosterone-induced 12- and 15-LO type 2 mRNA expressions were blocked by the EGF-receptor antagonist AG 1478 and by the MAPK-kinase inhibitor UO126. Aldosterone-treated VSMC also showed increased LDL oxidation, (approximately 2-fold; P < 0.001), which was blocked by spironolactone. In conclusion, aldosterone increased 12- and 15-LO expression in human VSMC, in association with increased 12- and 15-HETE generation and enhanced LDL oxidation and may directly augment VSMC contractility, hypertrophy, and migration through 12-HETE and promote LDL oxidation via the pro-oxidative properties of these enzymes.
Collapse
Affiliation(s)
- Rona Limor
- Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Aldosterone regulates blood pressure through its effects on the cardiovascular system and kidney. Aldosterone can also contribute to the development of hypertension that leads to chronic pathologies such as nephropathy and renal fibrosis. Aldosterone directly modulates renal cell proliferation and differentiation as part of normal kidney development. The stimulation of rapidly activated protein kinase cascades is one facet of how aldosterone regulates renal cell growth. These cascades may also contribute to myofibroblastic transformation and cell proliferation observed in pathological conditions of the kidney. Polycystic kidney disease is a genetic disorder that is accelerated by hypertension. EGFR-dependent proliferation of the renal epithelium is a factor in cyst development and trans-activation of EGFR is a key feature in initiating aldosterone-induced signalling cascades. Delineating the components of aldosterone-induced signalling cascades may identify novel therapeutic targets for proliferative diseases of the kidney.
Collapse
Affiliation(s)
- Warren Thomas
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland.
| | | | | |
Collapse
|
49
|
Krug AW, Allenhöfer L, Monticone R, Spinetti G, Gekle M, Wang M, Lakatta EG. Elevated mineralocorticoid receptor activity in aged rat vascular smooth muscle cells promotes a proinflammatory phenotype via extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase and epidermal growth factor receptor-dependent pathways. Hypertension 2010; 55:1476-83. [PMID: 20421514 DOI: 10.1161/hypertensionaha.109.148783] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Arterial aging is a predominant risk factor for the onset of cardiovascular diseases, such as hypertension, myocardial infarction, or stroke. Aging is associated with intravascular renin-angiotensin system activation, increased vascular stiffness, intima-media thickening, and a proinflammatory phenotype. Little is known about the influence of aldosterone on arterial aging. Hence, we hypothesized that aldosterone and mineralocorticoid receptor (MR) activation might contribute to and possibly accelerate the arterial aging process. We demonstrate increased MR expression in whole aortae and early passage aortic vascular smooth muscle cells from aged (30 months) compared with adult (8 months) F344XBN rats. Sensitivity to aldosterone-induced extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase activity is increased in aged cells. MR blockade and extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase inhibition prevent age-associated increases of transforming growth factor-beta, intercellular adhesion molecule 1, and procollagen 1. Aldosterone increases expression of proinflammatory marker proteins, shifting the phenotype of adult vascular smooth muscle cells toward the proinflammatory phenotype of aged rats. Epidermal growth factor receptor expression is increased with age and by aldosterone, and inhibition of epidermal growth factor receptor tyrosine kinase decreases age-associated proinflammatory marker expression. Our data support the hypothesis that increased constitutive MR signaling may promote and amplify age-associated inflammation that accompanies arterial aging through increased angiotensin II-stimulated expression of MR and enhanced sensitivity to aldosterone-mediated extracellular signal-regulated kinase 1/2 activation, likely related to increased epidermal growth factor receptor expression.
Collapse
Affiliation(s)
- Alexander W Krug
- National Institutes of Health, National Institute on Aging, Laboratory of Cardiovascular Science, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
Grossmann C, Wuttke M, Ruhs S, Seiferth A, Mildenberger S, Rabe S, Schwerdt G, Gekle M. Mineralocorticoid receptor inhibits CREB signaling by calcineurin activation. FASEB J 2010; 24:2010-9. [PMID: 20103717 DOI: 10.1096/fj.09-146985] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
We investigated the interaction of MR with cAMP-response element binding protein (CREB) and provide a mechanistic explanation and insights into the cellular relevance. MR --> CREB crosstalk was assessed in vascular smooth muscle cells and heterologous expression systems. Experiments were designed in a way that only one variable changed at a time and the respective vehicles served as controls. MR, but not GR, activation (aldosterone or hydrocortisone, IC(50), approximately 0.3 nM) inhibits CREB transcriptional activity induced by stimulation of beta1/2-adrenoceptors and adenylyl cyclase or addition of membrane-permeable cAMP up to 70% within 2 h after addition. The MR DNA-binding domain is not required for this inhibition. cAMP formation is virtually unchanged, whereas MR exerts a robust inhibition of CREB(S133) phosphorylation via calcineurin/PP2B activation without changes in PP2B-Aalpha or beta expression. In parallel, the PP2B-sensitive NFaT-pathway is activated. The inhibitory crosstalk attenuates CREB-induced glucose-6-phosphate dehydrogenase expression. Overall, transcriptional relevant MR --> CREB crosstalk occurs at the level of CREB phosphorylation by enhanced calcineurin activity, enables GRE-independent genomic signaling of MR, and is of potential pathophysiological relevance.
Collapse
Affiliation(s)
- Claudia Grossmann
- Julius-Bernstein-Institut für Physiologie, Universität Halle-Wittenberg, Halle, Germany
| | | | | | | | | | | | | | | |
Collapse
|