1
|
Cardiovascular Disease in Obstructive Sleep Apnea: Putative Contributions of Mineralocorticoid Receptors. Int J Mol Sci 2023; 24:ijms24032245. [PMID: 36768567 PMCID: PMC9916750 DOI: 10.3390/ijms24032245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/12/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a chronic and highly prevalent condition that is associated with oxidative stress, inflammation, and fibrosis, leading to endothelial dysfunction, arterial stiffness, and vascular insulin resistance, resulting in increased cardiovascular disease and overall mortality rates. To date, OSA remains vastly underdiagnosed and undertreated, with conventional treatments yielding relatively discouraging results for improving cardiovascular outcomes in OSA patients. As such, a better mechanistic understanding of OSA-associated cardiovascular disease (CVD) and the development of novel adjuvant therapeutic targets are critically needed. It is well-established that inappropriate mineralocorticoid receptor (MR) activation in cardiovascular tissues plays a causal role in a multitude of CVD states. Clinical studies and experimental models of OSA lead to increased secretion of the MR ligand aldosterone and excessive MR activation. Furthermore, MR activation has been associated with worsened OSA prognosis. Despite these documented relationships, there have been no studies exploring the causal involvement of MR signaling in OSA-associated CVD. Further, scarce clinical studies have exclusively assessed the beneficial role of MR antagonists for the treatment of systemic hypertension commonly associated with OSA. Here, we provide a comprehensive overview of overlapping mechanistic pathways recruited in the context of MR activation- and OSA-induced CVD and propose MR-targeted therapy as a potential avenue to abrogate the deleterious cardiovascular consequences of OSA.
Collapse
|
2
|
Abstract
Besides the physiological regulation of water, sodium, and potassium homeostasis, aldosterone modulates several physiological and pathological processes in the cardiovascular system. At the vascular level, aldosterone excess stimulates endothelial dysfunction and infiltration of inflammatory cells, enhances the development of the atherosclerotic plaque, and favors plaque instability, arterial stiffness, and calcification. At the cardiac level, aldosterone increases cardiac inflammation, fibrosis, and myocardial hypertrophy. As a clinical consequence, high aldosterone levels are associated with enhanced risk of cardiovascular events and mortality, especially when aldosterone secretion is inappropriate for renin levels and sodium intake, as in primary aldosteronism. Several clinical trials showed that mineralocorticoid receptor antagonists reduce cardiovascular mortality in patients with heart failure and reduced ejection fraction, but inconclusive results were reported for other cardiovascular conditions, such as heart failure with preserved ejection fraction, myocardial infarction, and atrial fibrillation. In patients with primary aldosteronism, adrenalectomy or treatment with mineralocorticoid receptor antagonists significantly mitigate adverse aldosterone effects, reducing the risk of cardiovascular events, mortality, and incident atrial fibrillation. In this review, we will summarize the major preclinical and clinical studies investigating the cardiovascular damage mediated by aldosterone and the protective effect of mineralocorticoid receptor antagonists for the reduction of cardiovascular risk in patients with cardiovascular diseases and primary aldosteronism.
Collapse
Affiliation(s)
- Fabrizio Buffolo
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Martina Tetti
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Silvia Monticone
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| |
Collapse
|
3
|
DeFronzo RA, Bakris GL. Modifying chronic kidney disease progression with the mineralocorticoid receptor antagonist finerenone in patients with type 2 diabetes. Diabetes Obes Metab 2022; 24:1197-1205. [PMID: 35302284 PMCID: PMC9323420 DOI: 10.1111/dom.14696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 11/29/2022]
Abstract
In patients with type 2 diabetes, chronic kidney disease (CKD) is the most common cause of kidney failure. With its increasing prevalence and limited treatment options, CKD is a major contributor to the global burden of disease. Although recent guidelines for the control of hypertension and hyperglycaemia, as well as the use of renin-angiotensin system inhibitors and, more recently, sodium-glucose co-transporter-2 inhibitors, have improved outcomes for patients with CKD and diabetes, there is still a high residual risk of CKD progression and adverse cardiovascular events. In this review, we discuss the recently published FIDELIO-DKD and FIGARO-DKD studies and FIDELITY prespecified individual patient analysis. Together, these studies have established finerenone, a novel non-steroidal mineralocorticoid receptor antagonist, as an effective treatment for kidney and cardiovascular protection and welcome addition to the pillars of treatment to slow CKD progression in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Ralph A. DeFronzo
- Diabetes Division, Dept of Medicine, UT Health and Texas Diabetes InstituteSan AntonioTexasUSA
| | - George L. Bakris
- University of Chicago School of Medicine, Dept of MedicineChicagoIllinoisUSA
| |
Collapse
|
4
|
Regulation of endothelial progenitor cell functions during hyperglycemia: new therapeutic targets in diabetic wound healing. J Mol Med (Berl) 2022; 100:485-498. [PMID: 34997250 DOI: 10.1007/s00109-021-02172-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/16/2021] [Accepted: 12/02/2021] [Indexed: 11/09/2022]
Abstract
Diabetes is primarily characterized by hyperglycemia, and its high incidence is often very costly to patients, their families, and national economies. Unsurprisingly, the number and function of endothelial progenitor cells (EPCs) decrease in patients resulting in diabetic wound non-healing. As precursors of endothelial cells (ECs), these cells were discovered in 1997 and found to play an essential role in wound healing. Their function, number, and role in wound healing has been widely investigated. Hitherto, a lot of complex molecular mechanisms have been discovered. In this review, we summarize the mechanisms of how hyperglycemia affects the function and number of EPCs and how the affected cells impact wound healing. We aim to provide a complete summary of the relationship between diabetic hyperglycosemia, EPCs, and wound healing, as well as a better comprehensive platform for subsequent related research.
Collapse
|
5
|
Kumboyono K, Chomsy IN, Nurwidyaningtyas W, Cesa FY, Tjahjono CT, Wihastuti TA. Differences in senescence of late Endothelial Progenitor Cells in non-smokers and smokers. Tob Induc Dis 2021; 19:10. [PMID: 34131419 PMCID: PMC8171388 DOI: 10.18332/tid/135320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/30/2021] [Accepted: 03/31/2021] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Endothelial Progenitor Cells (EPCs) are part of hematopoietic stem cells that differentiate into endothelial cells during their blood vessels' maturation process. The role of EPCs is widely known to contribute to repair of the vascular wall when endothelial dysfunction occurs. However, various risk factors for cardiovascular disease (CVD) influence EPC performance, leading to endothelial dysfunction. One EPC dysfunction is decreased amount of EPC mobilization to the injured tissue. EPC dysfunction reduces the angiogenetic function of EPCs. The vital maturation process that the EPCs must pass is the late phase. The dysfunction of late EPCs is known as senescence. This study aimed to identify and compare senescence of late EPCs, through CD62E and CD41 markers, in non-smokers and smokers as a risk factor for CVD. METHODS EPC collection was from peripheral mononuclear cells (PBMCs) in non-smokers (n=30) and smokers (n=31). The EPCs were then marked by CD62E/CD41 and senescence β-galactosidase assay using FACS. Identification of senescence cells was based on fluorescence with DAPI. RESULTS Positive percentage of late EPCs in non-smokers was not significantly different from that in smokers (p=0.014). The number of senescent late EPCs in smokers was higher than in non-smokers (p<0.0001). CONCLUSIONS Endothelial progenitor cells that experienced senescence in the smokers showed EPC dysfunction, which resulted in decreased cell angiogenic function. Further research is needed to explain the mechanism of re-endothelialization failure in EPC dysfunction due to smoking.
Collapse
Affiliation(s)
- Kumboyono Kumboyono
- School of Nursing, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | | | | | | | - Cholid Tri Tjahjono
- Department of Cardiology, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Titin Andri Wihastuti
- Department of Basic Nursing Science, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| |
Collapse
|
6
|
Gromotowicz-Poplawska A, Szoka P, Zakrzeska A, Kolodziejczyk P, Marcinczyk N, Szemraj J, Tutka P, Chabielska E. Hyperglycemia Potentiates Prothrombotic Effect of Aldosterone in a Rat Arterial Thrombosis Model. Cells 2021; 10:cells10020471. [PMID: 33671798 PMCID: PMC7927020 DOI: 10.3390/cells10020471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 11/16/2022] Open
Abstract
We investigated the role of aldosterone (ALDO) in the development of arterial thrombosis in streptozotocin-induced diabetic rats. To evaluate the effect of endogenous ALDO, the rats underwent adrenalectomy (ADX). ADX reduced the development of arterial thrombosis. A 1 h infusion of ALDO (30 μg/kg/h) enhanced thrombosis in adrenalectomized rats, while this effect was potentiated in diabetic rats. ALDO shortened bleeding time, increased plasma levels of tissue factor (TF) and plasminogen activator inhibitor, decreased plasma level of nitric oxide (NO) metabolites, and increased oxidative stress. Moreover, 2 h incubation of human umbilical vein endothelial cells (HUVECs) with ALDO (10-7 M) disrupted hemostatic balance in endothelial cells in normoglycemia (glucose 5.5 mM), and this effect was more pronounced in hyperglycemia (glucose 30 mM). We demonstrated that the acute ALDO infusion enhances arterial thrombosis in rats and hyperglycemia potentiates this prothrombotic effect. The mechanism of ALDO action was partially mediated by mineralocorticoid (MR) and glucocorticoid (GR) receptors and related to impact of the hormone on primary hemostasis, TF-dependent coagulation cascade, fibrinolysis, NO bioavailability, and oxidative stress balance. Our in vitro study confirmed that ALDO induces prothrombotic phenotype in the endothelium, particularly under hyperglycemic conditions.
Collapse
Affiliation(s)
- Anna Gromotowicz-Poplawska
- Department of Biopharmacy, Medical University of Bialystok, 15-222 Bialystok, Poland; (N.M.); (E.C.)
- Correspondence: ; Tel.: +48-857485804
| | - Piotr Szoka
- Department of Pharmacology, Medical University of Bialystok, 15-222 Bialystok, Poland;
| | | | - Patrycjusz Kolodziejczyk
- Department of Experimental and Clinical Pharmacology, University of Rzeszow, 35-959 Rzeszow, Poland; (P.K.); (P.T.)
| | - Natalia Marcinczyk
- Department of Biopharmacy, Medical University of Bialystok, 15-222 Bialystok, Poland; (N.M.); (E.C.)
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland;
| | - Piotr Tutka
- Department of Experimental and Clinical Pharmacology, University of Rzeszow, 35-959 Rzeszow, Poland; (P.K.); (P.T.)
- National Drug and Alcohol Research Center, University of New South Wales, Sydney 2052, Australia
| | - Ewa Chabielska
- Department of Biopharmacy, Medical University of Bialystok, 15-222 Bialystok, Poland; (N.M.); (E.C.)
| |
Collapse
|
7
|
Wang X, Luo Q, Wang M, Hu J, Zhang D, Zhang W, Wang G, Li N. Long-term impact of spironolactone compliance on microalbuminuria in patients with primary aldosteronism. Hypertens Res 2020; 44:426-434. [PMID: 33299114 DOI: 10.1038/s41440-020-00589-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/30/2020] [Accepted: 09/11/2020] [Indexed: 11/09/2022]
Abstract
Patients with primary aldosteronism (PA) have a high prevalence of microalbuminuria (MAU), which leads to more severe systemic vascular damage. However, the primary recommended drug treatment for PA, spironolactone (SPL), has had poor patient compliance owing to its adverse effects, and the effect of SPL compliance on MAU has not been fully evaluated in patients with PA. We analyzed the effect of SPL compliance on endothelial dysfunction by assessing MAU in patients with PA. The study included 145 confirmed PA patients who received long-term medical treatment (mean, 5 years). As expected, compliance with SPL treatment improved patients' blood pressure and serum potassium levels. Patients with PA who complied fully with SPL treatment had a lower rate of MAU than noncompliant patients (13.73% versus 34.88%, respectively; P = 0.004). Multivariate logistic regression analyses adjusted for age and sex showed that continuous SPL treatment was associated with a lower presence of MAU (odds ratio, 0.319; 95% confidence interval, 0.135-0.750; P = 0.009). This association remained significant after further adjusting for other major risk factors. However, in the subgroup analysis, the protective effect against MAU was limited in compliant patients treated with ≥40 mg/day SPL compared with noncompliant patients (9.62% versus 34.88%, respectively; P < 0.05). Our findings demonstrated that in addition to improving high blood pressure and hypokalemia, full compliance with the appropriate dose of SPL may benefit endothelial function, as reflected by a lower prevalence of MAU in patients with PA.
Collapse
Affiliation(s)
- Xiaotong Wang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region; Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, 830001, Xinjiang, China
| | - Qin Luo
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region; Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, 830001, Xinjiang, China
| | - Menghui Wang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region; Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, 830001, Xinjiang, China
| | - Junli Hu
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region; Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, 830001, Xinjiang, China
| | - Delian Zhang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region; Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, 830001, Xinjiang, China
| | - Weiwei Zhang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region; Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, 830001, Xinjiang, China
| | - Guoliang Wang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region; Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, 830001, Xinjiang, China
| | - Nanfang Li
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region; Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, 830001, Xinjiang, China.
| |
Collapse
|
8
|
Li N, Lin M, Heizhati M, Wang L, Luo Q, Li Y, Yili J, Hong J, Yao X, Zhu Q. Effect of spironolactone on cardiovascular morbidity and mortality in patients with hypertension and glucose metabolism disorders (ESCAM): a study protocol for a pragmatic randomised controlled trial. BMJ Open 2020; 10:e038694. [PMID: 33444188 PMCID: PMC7678363 DOI: 10.1136/bmjopen-2020-038694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Hypertension combined with diabetes and hypokalemia is more likely to develop hyperaldosteronism and is at higher risk of cardiovascular events. There is evidence that activation of aldosterone and mineralocorticoid receptors may play a significant role in the occurrence of cardiovascular events in patients with hypertension and diabetes. Clinical studies have demonstrated that spironolactone can reduce the incidence of cardiovascular events in patients with chronic kidney diseases or severe heart failure. However, the effect of spironolactone on cardiovascular risk in patients with hypertension and glucose metabolism disorders (GMD) and low potassium has been scarcely studied. Therefore, this study aims to evaluate whether add-on spironolactone (conventional antihypertensive drugs alone vs conventional antihypertensive drugs+spironolactone) can reduce the morbidity and mortality of cardiovascular events in this population. METHODS AND ANALYSIS In this multicentre, randomised, parallel-controlled study, a total of 7140 hypertensive patients aged 45-75 years with GMD and low potassium will be randomised in a 1:1 manner to the control or the spironolactone group (20 mg/day or with a maximum dose of 40 mg). The primary objective is to estimate the difference in the HR of composite cardiovascular events between the two groups. We will also assess the effects of spironolactone on individual cardiovascular events and the progression of diabetes and renal dysfunction. ETHICS AND DISSEMINATION This protocol was approved by the Independent Ethics Committee of People's Hospital of Xinjiang Uygur Autonomous Region (no. 2020020618). The results will be disseminated in peer-reviewed journals and at scientific conferences. TRIAL REGISTRATION NUMBER ChiCTR2000028909.
Collapse
Affiliation(s)
- Nanfang Li
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, China
| | - Mengyue Lin
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, China
- Xinjiang Medical University, Urumqi, China
| | - Mulalibieke Heizhati
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, China
| | - Lin Wang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, China
- Xinjiang Medical University, Urumqi, China
| | - Qin Luo
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, China
| | - Yuanyuan Li
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, China
| | - Jina Yili
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, China
| | - Jing Hong
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, China
| | - Xiaoguang Yao
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, China
| | - Qing Zhu
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, National Health Committee Key Laboratory of Hypertension Clinical Research, Urumqi, China
| |
Collapse
|
9
|
Liu J, Dou Q, Zhou C, Zhou L, Zhao F, Xu L, Xu Z, Ge Y, Wu R, Jia R. Low-energy shock wave pretreatment recruit circulating endothelial progenitor cells to attenuate renal ischaemia reperfusion injury. J Cell Mol Med 2020; 24:10589-10603. [PMID: 32761803 PMCID: PMC7521246 DOI: 10.1111/jcmm.15678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022] Open
Abstract
Low‐energy shock wave (LESW) has been recognized as a promising non‐invasive intervention to prevent the organs or tissues against ischaemia reperfusion injury (IRI), whereas its effect on kidney injury is rarely explored. To investigate the protective role of pretreatment with LESW on renal IRI in rats, animals were randomly divided into Sham, LESW, IRI and LESW + IRI groups. At 4, 12, 24 hours and 3 and 7 days after reperfusion, serum samples and renal tissues were harvested for performing the analysis of renal function, histopathology, immunohistochemistry, flow cytometry and Western blot, as well as enzyme‐linked immunosorbent assay. Moreover, circulating endothelial progenitor cells (EPCs) were isolated, labelled with fluorescent dye and injected by tail vein. The fluorescent signals of EPCs were detected using fluorescence microscope and in vivo imaging system to track the distribution of injected circulating EPCs. Results showed that pretreatment with LESW could significantly reduce kidney injury biomarkers, tubular damage, and cell apoptosis, and promote cell proliferation and vascularization in IRI kidneys. The renoprotective role of LESW pretreatment would be attributed to the remarkably increased EPCs in the treated kidneys, part of which were recruited from circulation through SDF‐1/CXCR7 pathway. In conclusion, pretreatment with LESW could increase the recruitment of circulating EPCs to attenuate and repair renal IRI.
Collapse
Affiliation(s)
- Jingyu Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Quanliang Dou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Changcheng Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Feng Zhao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zheng Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuzheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ran Wu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Lin L, Zhang L, Li XT, Ji JK, Chen XQ, Li YL, Li C. Rhynchophylline Attenuates Senescence of Endothelial Progenitor Cells by Enhancing Autophagy. Front Pharmacol 2020; 10:1617. [PMID: 32047439 PMCID: PMC6997466 DOI: 10.3389/fphar.2019.01617] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/11/2019] [Indexed: 12/25/2022] Open
Abstract
The increase of blood pressure accelerates endothelial progenitor cells (EPCs) senescence, hence a significant reduction in the number of EPCs is common in patients with hypertension. Autophagy is a defense and stress regulation mechanism to assist cell homeostasis and organelle renewal. A growing number of studies have found that autophagy has a positive role in repairing vascular injury, but the potential mechanism between autophagy and senescence of EPCs induced by hypertension has rarely been studied. Therefore, in this study, we aim to explore the relationship between senescence and autophagy, and investigate the protective effect of rhynchophylline (Rhy) on EPCs. In angiotensin II (Ang II)-treated EPCs, enhancing autophagy through rapamycin mitigated Ang II-induced cell senescence, on the contrary, 3-methyladenine aggravated the senescence by weakening autophagy. Similarly, Rhy attenuated senescence and improved cellular function by rescuing the impaired autophagy in Ang II-treated EPCs. Furthermore, we found that Rhy promoted autophagy by activating AMP-activated protein kinase (AMPK) signaling pathway. Our results show that enhanced autophagy attenuates EPCs senescence and Rhy rescues autophagy impairment to protect EPCs against Ang II injury.
Collapse
Affiliation(s)
- Lin Lin
- Institute of Traditional Chinese Medicine Innovation, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Zhang
- The First Faculty of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xin-Tong Li
- Institute of Education and Psychological Sciences, University of Jinan, Jinan, China
| | - Jing-Kang Ji
- Faculty of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Qing Chen
- Faculty of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yun-Lun Li
- Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- Institute of Traditional Chinese Medicine Innovation, Shandong University of Traditional Chinese Medicine, Jinan, China.,Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
11
|
Chen ZW, Tsai CH, Pan CT, Chou CH, Liao CW, Hung CS, Wu VC, Lin YH. Endothelial Dysfunction in Primary Aldosteronism. Int J Mol Sci 2019; 20:ijms20205214. [PMID: 31640178 PMCID: PMC6829211 DOI: 10.3390/ijms20205214] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Primary aldosteronism (PA) is characterized by excess production of aldosterone from the adrenal glands and is the most common and treatable cause of secondary hypertension. Aldosterone is a mineralocorticoid hormone that participates in the regulation of electrolyte balance, blood pressure, and tissue remodeling. The excess of aldosterone caused by PA results in an increase in cardiovascular and cerebrovascular complications, including coronary artery disease, myocardial infarction, stroke, transient ischemic attack, and even arrhythmia and heart failure. Endothelial dysfunction is a well-established fundamental cause of cardiovascular diseases and also a predictor of worse clinical outcomes. Accumulating evidence indicates that aldosterone plays an important role in the initiation and progression of endothelial dysfunction. Several mechanisms have been shown to contribute to aldosterone-induced endothelial dysfunction, including aldosterone-mediated vascular tone dysfunction, aldosterone- and endothelium-mediated vascular inflammation, aldosterone-related atherosclerosis, and vascular remodeling. These mechanisms are activated by aldosterone through genomic and nongenomic pathways in mineralocorticoid receptor-dependent and independent manners. In addition, other cells have also been shown to participate in these mechanisms. The complex interactions among endothelium, inflammatory cells, vascular smooth muscle cells and fibroblasts are crucial for aldosterone-mediated endothelial dysregulation. In this review, we discuss the association between aldosterone and endothelial function and the complex mechanisms from a molecular aspect. Furthermore, we also review current clinical research of endothelial dysfunction in patients with PA.
Collapse
Affiliation(s)
- Zheng-Wei Chen
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan.
- Cardiovascular center, National Taiwan University Hospital, Taipei 10002, Taiwan.
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin 64041, Taiwan.
| | - Cheng-Hsuan Tsai
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan.
- Cardiovascular center, National Taiwan University Hospital, Taipei 10002, Taiwan.
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Jin-Shan Branch, New Taipei City 20844, Taiwan.
| | - Chien-Ting Pan
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan.
- Cardiovascular center, National Taiwan University Hospital, Taipei 10002, Taiwan.
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin 64041, Taiwan.
| | - Chia-Hung Chou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10041, Taiwan.
| | - Che-Wei Liao
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu 30059, Taiwan.
| | - Chi-Sheng Hung
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan.
- Cardiovascular center, National Taiwan University Hospital, Taipei 10002, Taiwan.
| | - Vin-Cent Wu
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan.
| | - Yen-Hung Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 10002, Taiwan.
- Cardiovascular center, National Taiwan University Hospital, Taipei 10002, Taiwan.
| |
Collapse
|
12
|
Zhang R, Liu J, Yu S, Sun D, Wang X, Fu J, Shen J, Xie Z. Osteoprotegerin (OPG) Promotes Recruitment of Endothelial Progenitor Cells (EPCs) via CXCR4 Signaling Pathway to Improve Bone Defect Repair. Med Sci Monit 2019; 25:5572-5579. [PMID: 31350844 PMCID: PMC6681686 DOI: 10.12659/msm.916838] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/01/2019] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The aim of this study was to investigate the effect of using osteoprotegerin (OPG) to treat bone defects mediated by endothelial progenitor cell (EPC) recruitment and migration through the CXCR4 signaling pathway. MATERIAL AND METHODS The EPCs extracted from human peripheral blood were cultured in vitro and the expression of CXCR4 and its downstream p-AKT was monitored by the Western blot analysis after OPG treatment. Using the scratch wound healing test and Transwell assay, we assessed the variables influencing the effect of OPG on EPCs after pre-treatment with CXCR4 blocker (AMD3100) and PI3K blocker (Ly294002). After 4 weeks, the bone defect repair condition was estimated via micro-CT and staining with HE and Masson trichrome. Then, immunofluorescence staining was performed to assess angiogenesis in bone defects, while the expression of EPC marker and vascular endothelial growth factor receptor 2 (VEGFR2) was detected by immunohistochemical staining. RESULTS The EPCs treated with OPG had increased levels of CXCR4 and p-AKT. Moreover, the difference in EPC levels among groups in the scratch wound healing experiment and migration experiment indicated that the OPG treatment promoted cell migration and AMD3100 and LY294002 inhibited the function of OPG. In addition, OPG promoted angiogenesis and repair of bone defect in rats, and these effects were abolished by AMD3100 and LY294002 administration. CONCLUSIONS OPG enhanced the proliferation and migration of EPCs through the CXCR4 pathway and promoted angiogenesis and bone formation at bone defect sites.
Collapse
Affiliation(s)
- Rongfeng Zhang
- National and Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Jianwei Liu
- Department of Orthopedics, The First People’s Hospital of Chenzhou, Chenzhou, Hunan, P.R. China
| | - Shengpeng Yu
- National and Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Dong Sun
- National and Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Xiaohua Wang
- National and Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Jingshu Fu
- National and Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Jie Shen
- National and Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Zhao Xie
- National and Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| |
Collapse
|
13
|
Coats AJS. Figures of the Heart Failure Association: Professor Dr. med. Johann Bauersachs, Chair of the Clinical Science Section. Eur J Heart Fail 2019; 21:545-548. [PMID: 31069912 DOI: 10.1002/ejhf.1484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 11/05/2022] Open
|
14
|
Lother A, Deng L, Huck M, Fürst D, Kowalski J, Esser JS, Moser M, Bode C, Hein L. Endothelial cell mineralocorticoid receptors oppose VEGF-induced gene expression and angiogenesis. J Endocrinol 2019; 240:15-26. [PMID: 30400069 DOI: 10.1530/joe-18-0494] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 09/24/2018] [Indexed: 12/29/2022]
Abstract
Aldosterone is a key factor in adverse cardiovascular remodeling by acting on the mineralocorticoid receptor (MR) in different cell types. Endothelial MR activation mediates hypertrophy, inflammation and fibrosis. Cardiovascular remodeling is often accompanied by impaired angiogenesis, which is a risk factor for the development of heart failure. In this study, we evaluated the impact of MR in endothelial cells on angiogenesis. Deoxycorticosterone acetate (DOCA)-induced hypertension was associated with capillary rarefaction in the heart of WT mice but not of mice with cell type-specific MR deletion in endothelial cells. Consistently, endothelial MR deletion prevented the inhibitory effect of aldosterone on the capillarization of subcutaneously implanted silicon tubes and on capillary sprouting from aortic ring segments. We examined MR-dependent gene expression in cultured endothelial cells by RNA-seq and identified a cluster of differentially regulated genes related to angiogenesis. We found opposing effects on gene expression when comparing activation of the mineralocorticoid receptor in ECs to treatment with vascular endothelial growth factor (VEGF), a potent activator of angiogenesis. In conclusion, we demonstrate here that activation of endothelial cell MR impaired angiogenic capacity and lead to capillary rarefaction in a mouse model of MR-driven hypertension. MR activation opposed VEGF-induced gene expression leading to the dysregulation of angiogenesis-related gene networks in endothelial cells. Our findings underscore the pivotal role of endothelial cell MR in the pathophysiology of hypertension and related heart disease.
Collapse
Affiliation(s)
- Achim Lother
- A Lother, Institute of experimental and clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Lisa Deng
- L Deng, Institute of experimental and clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Michael Huck
- M Huck, Institute of experimental and clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - David Fürst
- D Fürst, Institute of experimental and clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Jessica Kowalski
- J Kowalski, Institute of experimental and clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Jennifer Susanne Esser
- J Esser, Heart Center, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Martin Moser
- M Moser, Heart Center, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- C Bode, Heart Center, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Lutz Hein
- L Hein, Institute of experimental and clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
15
|
González-Blázquez R, Somoza B, Gil-Ortega M, Martín Ramos M, Ramiro-Cortijo D, Vega-Martín E, Schulz A, Ruilope LM, Kolkhof P, Kreutz R, Fernández-Alfonso MS. Finerenone Attenuates Endothelial Dysfunction and Albuminuria in a Chronic Kidney Disease Model by a Reduction in Oxidative Stress. Front Pharmacol 2018; 9:1131. [PMID: 30356804 PMCID: PMC6189469 DOI: 10.3389/fphar.2018.01131] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/18/2018] [Indexed: 01/01/2023] Open
Abstract
Albuminuria is an early marker of renovascular damage associated to an increase in oxidative stress. The Munich Wistar Frömter (MWF) rat is a model of chronic kidney disease (CKD), which exhibits endothelial dysfunction associated to low nitric oxide availability. We hypothesize that the new highly selective, non-steroidal mineralocorticoid receptor (MR) antagonist, finerenone, reverses both endothelial dysfunction and microalbuminuria. Twelve-week-old MWF (MWF-C; MWF-FIN) and aged-matched normoalbuminuric Wistar (W-C; W-FIN) rats were treated with finerenone (FIN, 10 mg/kg/day p.o.) or vehicle (C) for 4-week. Systolic blood pressure (SBP) and albuminuria were determined the last day of treatment. Finerenone lowered albuminuria by >40% and significantly reduced SBP in MWF. Aortic rings of MWF-C showed higher contractions to either noradrenaline (NA) or angiotensin II (Ang II), and lower relaxation to acetylcholine (Ach) than W-C rings. These alterations were reversed by finerenone to W-C control levels due to an upregulation in phosphorylated Akt and eNOS, and an increase in NO availability. Apocynin and 3-amino-1,2,4-triazole significantly reduced contractions to NA or Ang II in MWF-C, but not in MWF-FIN rings. Accordingly, a significant increase of Mn-superoxide dismutase (SOD) and Cu/Zn-SOD protein levels were observed in rings of MWF-FIN, without differences in p22phox, p47phox or catalase levels. Total SOD activity was increased in kidneys from MWF-FIN rats. In conclusion, finerenone improves endothelial dysfunction through an enhancement in NO bioavailability and a decrease in superoxide anion levels due to an upregulation in SOD activity. This is associated with an increase in renal SOD activity and a reduction of albuminuria.
Collapse
Affiliation(s)
- Raquel González-Blázquez
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, Madrid, Spain
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, Madrid, Spain
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, Madrid, Spain
| | - Miriam Martín Ramos
- Instituto Pluridisciplinar and Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Elena Vega-Martín
- Instituto Pluridisciplinar and Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Angela Schulz
- Department of Clinical Pharmacology and Toxicology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Luis Miguel Ruilope
- Unidad de Hipertensión, Instituto de Investigación Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain.,Departamento de Medicina Preventiva y Salud Pública, Universidad Autónoma de Madrid, Madrid, Spain
| | - Peter Kolkhof
- Drug Discovery, Pharmaceuticals, Cardiology Research, Bayer HealthCare AG, Wuppertal, Germany
| | - Reinhold Kreutz
- Department of Clinical Pharmacology and Toxicology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - María S Fernández-Alfonso
- Instituto Pluridisciplinar and Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
16
|
Carolina E, Kato T, Khanh VC, Moriguchi K, Yamashita T, Takeuchi K, Hamada H, Ohneda O. Glucocorticoid Impaired the Wound Healing Ability of Endothelial Progenitor Cells by Reducing the Expression of CXCR4 in the PGE2 Pathway. Front Med (Lausanne) 2018; 5:276. [PMID: 30324106 PMCID: PMC6173212 DOI: 10.3389/fmed.2018.00276] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 09/10/2018] [Indexed: 01/05/2023] Open
Abstract
Background: Endothelial progenitor cells (EPCs) can be used to treat ischemic disease in cell-based therapy owing to their neovascularization potential. Glucocorticoids (GCs) have been widely used as strong anti-inflammatory reagents. However, despite their beneficial effects, side effects, such as impairing wound healing are commonly reported with GC-based therapy, and the effects of GC therapy on the wound healing function of EPCs are unclear. Methods: In this study, we investigated how GC treatment affects the characteristics and wound healing function of EPCs. Results: We found that GC treatment reduced the proliferative ability of EPCs. In addition, the expression of CXCR4 was dramatically impaired, which suppressed the migration of EPCs. A transplantation study in a flap mouse model revealed that GC-treated EPCs showed a poor homing ability to injured sites and a low activity for recruiting inflammatory cells, which led to wound healing dysfunction. Impairment of prostaglandin E2 (PGE2) synthases, cyclooxygenase (COX2) and microsomal PGE2 synthase 1 (mPEGS1) were identified as being involved in the GC-induced impairment of the CXCR4 expression in EPCs. Treatment with PGE2 rescued the expression of CXCR4 and restored the migration ability of GC-treated EPCs. In addition, the PGE2 signal that activated the PI3K/AKT pathway was identified to be involved in the regulation of CXCR4 in EPCs under the effects of GCs. In addition, similar negative effects of GCs were observed in EPCs under hypoxic conditions. Under hypoxic conditions, GCs independently impaired the PGE2 and HIF2α pathways, which downregulated the expression of CXCR4 in EPCs. Our findings highlighted the influences of GCs on the characteristics and functions of EPCs, suggesting that the use of EPCs for autologous cell transplantation in patients who have used GCs for a long time should be considered carefully.
Collapse
Affiliation(s)
- Erica Carolina
- Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Toshiki Kato
- Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan.,Ph.D. Program in Human Biology, School of Integrative Global Majors, University of Tsukuba, Tsukuba, Japan
| | - Vuong Cat Khanh
- Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Kana Moriguchi
- Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Toshiharu Yamashita
- Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Kosuke Takeuchi
- Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Hiromi Hamada
- Department of Obstetrics and Gynecology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Osamu Ohneda
- Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
17
|
The endothelial mineralocorticoid receptor: mediator of the switch from vascular health to disease. Curr Opin Nephrol Hypertens 2018; 26:97-104. [PMID: 27930384 DOI: 10.1097/mnh.0000000000000306] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW Endothelial dysfunction is an early feature of vascular disease induced by cardiovascular risk factors (CRFs). In growing populations with obesity, diabetes, hypertension, and heart failure, mineralocorticoid receptor antagonism improves endothelial function. This review summarizes recent advances in our understanding of the specific role of endothelial cell mineralocorticoid receptor in vascular function in health and disease. RECENT FINDINGS Using transgenic mice with mineralocorticoid receptor expression specifically modulated in endothelial cells, recent studies support the emerging concept that while endothelial cell mineralocorticoid receptor may be protective in health, in the presence of CRFs, endothelial cell mineralocorticoid receptor activity contributes to endothelial dysfunction and progression of vascular disease. Proposed mechanisms include a role for endothelial cell mineralocorticoid receptor in decreased nitric oxide production and bioavailability, increased vascular oxidative stress, regulation of epithelial sodium channels that enhance vascular stiffness, and increased endothelial cell adhesion molecules promoting inflammation. The role of endothelial cell mineralocorticoid receptor may also depend on the sex, race, or vascular bed involved. SUMMARY Recent advances support the idea that endothelial cell mineralocorticoid receptor is a mediator of the switch from vascular health to disease in response to CRFs. Further investigation of the molecular mechanism is underway to identify therapeutic interventions that will limit the detrimental effects of endothelial cell mineralocorticoid receptor in patients at cardiovascular risk.
Collapse
|
18
|
PR interval prolongation in coronary patients or risk equivalent: excess risk of ischemic stroke and vascular pathophysiological insights. BMC Cardiovasc Disord 2017; 17:233. [PMID: 28836952 PMCID: PMC5571504 DOI: 10.1186/s12872-017-0667-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/17/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Whether PR prolongation independently predicts new-onset ischemic events of myocardial infarction and stroke was unclear. Underlying pathophysiological mechanisms of PR prolongation leading to adverse cardiovascular events were poorly understood. We investigated the role of PR prolongation in pathophysiologically-related adverse cardiovascular events and underlying mechanisms. METHODS We prospectively investigated 597 high-risk cardiovascular outpatients (mean age 66 ± 11 yrs.; male 67%; coronary disease 55%, stroke 22%, diabetes 52%) for new-onset ischemic stroke, myocardial infarction (MI), congestive heart failure (CHF), and cardiovascular death. Vascular phenotype was determined by carotid intima-media thickness (IMT). RESULTS PR prolongation >200 ms was present in 79 patients (13%) at baseline. PR prolongation >200 ms was associated with significantly higher mean carotid IMT (1.05 ± 0.37 mm vs 0.94 ± 0.28 mm, P = 0.010). After mean study period of 63 ± 11 months, increased PR interval significantly predicted new-onset ischemic stroke (P = 0.006), CHF (P = 0.040), cardiovascular death (P < 0.001), and combined cardiovascular endpoints (P < 0.001) at cut-off >200 ms. Using multivariable Cox regression, PR prolongation >200 ms independently predicted new-onset ischemic stroke (HR 8.6, 95% CI: 1.9-37.8, P = 0.005), cardiovascular death (HR 14.1, 95% CI: 3.8-51.4, P < 0.001) and combined cardiovascular endpoints (HR 2.4, 95% CI: 1.30-4.43, P = 0.005). PR interval predicts new-onset MI at the exploratory cut-off >162 ms (C-statistic 0.70, P = 0.001; HR: 8.0, 95% CI: 1.65-38.85, P = 0.010). CONCLUSIONS PR prolongation strongly predicts new-onset ischemic stroke, MI, cardiovascular death, and combined cardiovascular endpoint including CHF in coronary patients or risk equivalent. Adverse vascular function may implicate an intermediate pathophysiological phenotype or mediating mechanism.
Collapse
|
19
|
Endothelial progenitor cells and hypertension: current concepts and future implications. Clin Sci (Lond) 2017; 130:2029-2042. [PMID: 27729472 DOI: 10.1042/cs20160587] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/09/2016] [Indexed: 02/07/2023]
Abstract
The discovery of endothelial progenitor cells (EPCs), a group of cells that play important roles in angiogenesis and the maintenance of vascular endothelial integrity, has led to considerable improvements in our understanding of the circulatory system and the regulatory mechanisms of vascular homoeostasis. Despite lingering disputes over where EPCs actually originate and how they facilitate angiogenesis, extensive research in the past decade has brought about significant advancements in this field of research, establishing EPCs as an essential element in the pathogenesis of various diseases. EPC and hypertensive disorders, especially essential hypertension (EH, also known as primary hypertension), represent one of the most appealing branches in this area of research. Chronic hypertension remains a major threat to public health, and the exact pathologic mechanisms of EH have never been fully elucidated. Is there a relationship between EPC and hypertension? If so, what is the nature of such relationship-is it mediated by blood pressure alterations, or other factors that lie in between? How can our current knowledge about EPCs be utilized to advance the prevention and clinical management of hypertension? In this review, we set out to answer these questions by summarizing the current concepts about EPC pathophysiology in the context of hypertension, while attempting to point out directions for future research on this subject.
Collapse
|
20
|
Guo Y, Ledesma RA, Peng R, Liu Q, Xu D. The Beneficial Effects of Cardiac Rehabilitation on the Function and Levels of Endothelial Progenitor Cells. Heart Lung Circ 2016; 26:10-17. [PMID: 27614559 DOI: 10.1016/j.hlc.2016.06.1210] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 04/08/2016] [Accepted: 06/06/2016] [Indexed: 12/19/2022]
Abstract
Cardiac rehabilitation (CR) is a comprehensive program, which mainly focusses on exercise training, disease evaluation, cardiovascular risk factors control, medication therapy, psychosocial intervention, and patient education. Although the beneficial properties of CR have been widely evidenced, its mechanism is still not completely clarified. To date, endothelial progenitor cells (EPCs) have been explored by emerging studies, and evidence has suggested that CR, especially exercise training, significantly increases the function and levels of EPCs, which is likely to elucidate the profiting mechanism of CR. Thus, this review summarises the potential relationship between CR and EPCs with an aim of providing novel directions for future CR research.
Collapse
Affiliation(s)
- Yuan Guo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Robert Andre Ledesma
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, USA
| | - Ran Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qiong Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
21
|
Chen L, Ding ML, Wu F, He W, Li J, Zhang XY, Xie WL, Duan SZ, Xia WH, Tao J. Impaired Endothelial Repair Capacity of Early Endothelial Progenitor Cells in Hypertensive Patients With Primary Hyperaldosteronemia: Role of 5,6,7,8-Tetrahydrobiopterin Oxidation and Endothelial Nitric Oxide Synthase Uncoupling. Hypertension 2016; 67:430-9. [PMID: 26628675 DOI: 10.1161/hypertensionaha.115.06597] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 11/08/2015] [Indexed: 01/22/2023]
Abstract
Although hyperaldosteronemia exerts detrimental impacts on vascular endothelium in addition to elevating blood pressure, the effects and molecular mechanisms of hyperaldosteronemia on early endothelial progenitor cell (EPC)-mediated endothelial repair after arterial damage are yet to be determined. The aim of this study was to investigate the endothelial repair capacity of early EPCs from hypertensive patients with primary hyperaldosteronemia (PHA). In vivo endothelial repair capacity of early EPCs from PHAs (n=20), age- and blood pressure-matched essential hypertension patients (n=20), and age-matched healthy subjects (n=20) was evaluated by transplantation into a nude mouse carotid endothelial denudation model. Endothelial function was evaluated by flow-mediated dilation of brachial artery in human subjects. In vivo endothelial repair capacity of early EPCs and flow-mediated dilation were impaired both in PHAs and in essential hypertension patients when compared with age-matched healthy subjects; however, the early EPC in vivo endothelial repair capacity and flow-mediated dilation of PHAs were impaired more severely than essential hypertension patients. Oral spironolactone improved early EPC in vivo endothelial repair capacity and flow-mediated dilation of PHAs. Increased oxidative stress, oxidative 5,6,7,8-tetrahydrobiopterin degradation, endothelial nitric oxide synthase uncoupling and decreased nitric oxide production were found in early EPCs from PHAs. Nicotinamide adenine dinucleotide phosphate oxidase subunit p47(phox) knockdown or 5,6,7,8-tetrahydrobiopterin supplementation attenuated endothelial nitric oxide synthase uncoupling and enhanced in vivo endothelial repair capacity of early EPCs from PHAs. In conclusion, PHAs exhibited more impaired endothelial repair capacity of early EPCs than did essential hypertension patients independent of blood pressure, which was associated with mineralocorticoid receptor-dependent oxidative stress and subsequently 5,6,7,8-tetrahydrobiopterin degradation and endothelial nitric oxide synthase uncoupling.
Collapse
Affiliation(s)
- Long Chen
- From the Department of Hypertension and Vascular Disease (L.C., F.W., X.-Y.Z., W.-L.X., W.-H.X., J.T.), Department of Geriatrics (L.C., M.-L.D., F.W., W.H., J.L., J.T.), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (S.-Z.D.)
| | - Mei-Lin Ding
- From the Department of Hypertension and Vascular Disease (L.C., F.W., X.-Y.Z., W.-L.X., W.-H.X., J.T.), Department of Geriatrics (L.C., M.-L.D., F.W., W.H., J.L., J.T.), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (S.-Z.D.)
| | - Fang Wu
- From the Department of Hypertension and Vascular Disease (L.C., F.W., X.-Y.Z., W.-L.X., W.-H.X., J.T.), Department of Geriatrics (L.C., M.-L.D., F.W., W.H., J.L., J.T.), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (S.-Z.D.)
| | - Wen He
- From the Department of Hypertension and Vascular Disease (L.C., F.W., X.-Y.Z., W.-L.X., W.-H.X., J.T.), Department of Geriatrics (L.C., M.-L.D., F.W., W.H., J.L., J.T.), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (S.-Z.D.)
| | - Jin Li
- From the Department of Hypertension and Vascular Disease (L.C., F.W., X.-Y.Z., W.-L.X., W.-H.X., J.T.), Department of Geriatrics (L.C., M.-L.D., F.W., W.H., J.L., J.T.), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (S.-Z.D.)
| | - Xiao-Yu Zhang
- From the Department of Hypertension and Vascular Disease (L.C., F.W., X.-Y.Z., W.-L.X., W.-H.X., J.T.), Department of Geriatrics (L.C., M.-L.D., F.W., W.H., J.L., J.T.), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (S.-Z.D.)
| | - Wen-Li Xie
- From the Department of Hypertension and Vascular Disease (L.C., F.W., X.-Y.Z., W.-L.X., W.-H.X., J.T.), Department of Geriatrics (L.C., M.-L.D., F.W., W.H., J.L., J.T.), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (S.-Z.D.)
| | - Sheng-Zhong Duan
- From the Department of Hypertension and Vascular Disease (L.C., F.W., X.-Y.Z., W.-L.X., W.-H.X., J.T.), Department of Geriatrics (L.C., M.-L.D., F.W., W.H., J.L., J.T.), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (S.-Z.D.)
| | - Wen-Hao Xia
- From the Department of Hypertension and Vascular Disease (L.C., F.W., X.-Y.Z., W.-L.X., W.-H.X., J.T.), Department of Geriatrics (L.C., M.-L.D., F.W., W.H., J.L., J.T.), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (S.-Z.D.).
| | - Jun Tao
- From the Department of Hypertension and Vascular Disease (L.C., F.W., X.-Y.Z., W.-L.X., W.-H.X., J.T.), Department of Geriatrics (L.C., M.-L.D., F.W., W.H., J.L., J.T.), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, China (S.-Z.D.).
| |
Collapse
|
22
|
Finan A, Richard S. Stimulating endogenous cardiac repair. Front Cell Dev Biol 2015; 3:57. [PMID: 26484341 PMCID: PMC4586501 DOI: 10.3389/fcell.2015.00057] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 09/08/2015] [Indexed: 01/10/2023] Open
Abstract
The healthy adult heart has a low turnover of cardiac myocytes. The renewal capacity, however, is augmented after cardiac injury. Participants in cardiac regeneration include cardiac myocytes themselves, cardiac progenitor cells, and peripheral stem cells, particularly from the bone marrow compartment. Cardiac progenitor cells and bone marrow stem cells are augmented after cardiac injury, migrate to the myocardium, and support regeneration. Depletion studies of these populations have demonstrated their necessary role in cardiac repair. However, the potential of these cells to completely regenerate the heart is limited. Efforts are now being focused on ways to augment these natural pathways to improve cardiac healing, primarily after ischemic injury but in other cardiac pathologies as well. Cell and gene therapy or pharmacological interventions are proposed mechanisms. Cell therapy has demonstrated modest results and has passed into clinical trials. However, the beneficial effects of cell therapy have primarily been their ability to produce paracrine effects on the cardiac tissue and recruit endogenous stem cell populations as opposed to direct cardiac regeneration. Gene therapy efforts have focused on prolonging or reactivating natural signaling pathways. Positive results have been demonstrated to activate the endogenous stem cell populations and are currently being tested in clinical trials. A potential new avenue may be to refine pharmacological treatments that are currently in place in the clinic. Evidence is mounting that drugs such as statins or beta blockers may alter endogenous stem cell activity. Understanding the effects of these drugs on stem cell repair while keeping in mind their primary function may strike a balance in myocardial healing. To maximize endogenous cardiac regeneration, a combination of these approaches could ameliorate the overall repair process to incorporate the participation of multiple cellular players.
Collapse
Affiliation(s)
- Amanda Finan
- Centre National de la Recherche Scientifique United Medical Resource 9214, Institut National de la Santé et de la Recherche Médicale U1046, Physiology and Experimental Medicine of the Heart and Muscles, University of Montpellier Montpellier, France
| | - Sylvain Richard
- Centre National de la Recherche Scientifique United Medical Resource 9214, Institut National de la Santé et de la Recherche Médicale U1046, Physiology and Experimental Medicine of the Heart and Muscles, University of Montpellier Montpellier, France
| |
Collapse
|
23
|
Könemann S, Wenzel K, Ameling S, Grube K, Hammer E, Könemann R, Samal R, Völker U, Felix SB. The Other Side of the RAAS: Aldosterone Improves Migration of Cardiac Progenitor Cells. J Cell Physiol 2015; 230:2829-36. [DOI: 10.1002/jcp.25013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 04/03/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Stephanie Könemann
- Department of Internal Medicine B; University Medicine Greifswald; Greifswald Germany
- DZHK (German Center for Cardiovascular Research); partner site Greifswald
| | - Kristin Wenzel
- Department of Internal Medicine B; University Medicine Greifswald; Greifswald Germany
- DZHK (German Center for Cardiovascular Research); partner site Greifswald
| | - Sabine Ameling
- DZHK (German Center for Cardiovascular Research); partner site Greifswald
- Interfaculty Institute for Genetic and Functional Genomics; University Medicine Greifswald; Greifswald Germany
| | - Karina Grube
- Department of Internal Medicine B; University Medicine Greifswald; Greifswald Germany
- DZHK (German Center for Cardiovascular Research); partner site Greifswald
| | - Elke Hammer
- DZHK (German Center for Cardiovascular Research); partner site Greifswald
- Interfaculty Institute for Genetic and Functional Genomics; University Medicine Greifswald; Greifswald Germany
| | - Raik Könemann
- Department of Internal Medicine B; University Medicine Greifswald; Greifswald Germany
| | - Rasmita Samal
- Interfaculty Institute for Genetic and Functional Genomics; University Medicine Greifswald; Greifswald Germany
| | - Uwe Völker
- DZHK (German Center for Cardiovascular Research); partner site Greifswald
- Interfaculty Institute for Genetic and Functional Genomics; University Medicine Greifswald; Greifswald Germany
| | - Stephan B. Felix
- Department of Internal Medicine B; University Medicine Greifswald; Greifswald Germany
- DZHK (German Center for Cardiovascular Research); partner site Greifswald
| |
Collapse
|
24
|
Vascular mineralocorticoid receptor and blood pressure regulation. Curr Opin Pharmacol 2015; 21:138-44. [DOI: 10.1016/j.coph.2015.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 01/16/2023]
|
25
|
Gravez B, Tarjus A, Pelloux V, Ouvrard‐Pascaud A, Delcayre C, Samuel J, Clément K, Farman N, Jaisser F, Messaoudi S. Aldosterone promotes cardiac endothelial cell proliferation in vivo. J Am Heart Assoc 2015; 4:e001266. [PMID: 25564371 PMCID: PMC4330055 DOI: 10.1161/jaha.114.001266] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 11/20/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND Experimentally, aldosterone in association with NaCl induces cardiac fibrosis, oxidative stress, and inflammation through mineralocorticoid receptor activation; however, the biological processes regulated by aldosterone alone in the heart remain to be identified. METHODS AND RESULTS Mice were treated for 7 days with aldosterone, and then cardiac transcriptome was analyzed. Aldosterone regulated 60 transcripts (51 upregulated and 9 downregulated) in the heart (fold change ≥1.5, false discovery rate <0.01). To identify the biological processes modulated by aldosterone, a gene ontology analysis was performed. The majority of aldosterone-regulated genes were involved in cell division. The cardiac Ki-67 index (an index of proliferation) of aldosterone-treated mice was higher than that of nontreated mice, confirming microarray predictions. Costaining of Ki-67 with vinculin, CD68, α-smooth muscle actin, CD31, or caveolin 1 revealed that the cycling cells were essentially endothelial cells. Aldosterone-induced mineralocorticoid receptor-dependent proliferation was confirmed ex vivo in human endothelial cells. Moreover, pharmacological-specific blockade of mineralocorticoid receptor by eplerenone inhibited endothelial cell proliferation in a preclinical model of heart failure (transverse aortic constriction). CONCLUSIONS Aldosterone modulates cardiac gene expression and induces the proliferation of cardiac endothelial cells in vivo.
Collapse
Affiliation(s)
- Basile Gravez
- Inserm U1138, Team 1, 15 rue de l'école de médecine, Paris, France (B.G., A.T., N.F., F.J., S.M.)
| | - Antoine Tarjus
- Inserm U1138, Team 1, 15 rue de l'école de médecine, Paris, France (B.G., A.T., N.F., F.J., S.M.)
| | - Véronique Pelloux
- INSERM‐UMR 1166 Team 6‐ GH Pitié‐Salpêtrière, 83 Bd de l'hôpital, Paris, France (P., K.C.)
| | | | - Claude Delcayre
- Inserm U942, 41 Boulevard de la chapelle, Paris, France (C.D., J.S.)
| | - Janelise Samuel
- Inserm U942, 41 Boulevard de la chapelle, Paris, France (C.D., J.S.)
| | - Karine Clément
- INSERM‐UMR 1166 Team 6‐ GH Pitié‐Salpêtrière, 83 Bd de l'hôpital, Paris, France (P., K.C.)
| | - Nicolette Farman
- Inserm U1138, Team 1, 15 rue de l'école de médecine, Paris, France (B.G., A.T., N.F., F.J., S.M.)
| | - Fréderic Jaisser
- Inserm U1138, Team 1, 15 rue de l'école de médecine, Paris, France (B.G., A.T., N.F., F.J., S.M.)
| | - Smail Messaoudi
- Inserm U1138, Team 1, 15 rue de l'école de médecine, Paris, France (B.G., A.T., N.F., F.J., S.M.)
| |
Collapse
|
26
|
Lother A, Moser M, Bode C, Feldman RD, Hein L. Mineralocorticoids in the heart and vasculature: new insights for old hormones. Annu Rev Pharmacol Toxicol 2014; 55:289-312. [PMID: 25251996 DOI: 10.1146/annurev-pharmtox-010814-124302] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The mineralocorticoid aldosterone is a key regulator of water and electrolyte homeostasis. Numerous recent developments have advanced the field of mineralocorticoid pharmacology—namely, clinical trials have shown the beneficial effects of aldosterone antagonists in chronic heart failure and post-myocardial infarction treatment. Experimental studies using cell type-specific gene targeting of the mineralocorticoid receptor (MR) gene in mice have revealed the importance of extrarenal aldosterone signaling in cardiac myocytes, endothelial cells, vascular smooth cells, and macrophages. In addition, several molecular pathways involving signal transduction via the classical MR as well as the G protein-coupled receptor GPER mediate the diverse spectrum of effects of aldosterone on cells. This knowledge has initiated the development of new pharmacological ligands to specifically interfere with targets on different levels of aldosterone signaling. For example, aldosterone synthase inhibitors such as LCI699 and the novel nonsteroidal MR antagonist BAY 94-8862 have been tested in clinical trials. Interference with the interaction between MR and its coregulators seems to be a promising strategy toward the development of selective MR modulators.
Collapse
Affiliation(s)
- Achim Lother
- Heart Center, Department of Cardiology and Angiology I, University of Freiburg, 79106 Freiburg, Germany;
| | | | | | | | | |
Collapse
|
27
|
Maron BA, Leopold JA. The role of the renin-angiotensin-aldosterone system in the pathobiology of pulmonary arterial hypertension (2013 Grover Conference series). Pulm Circ 2014; 4:200-10. [PMID: 25006439 PMCID: PMC4070776 DOI: 10.1086/675984] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 01/17/2014] [Indexed: 12/20/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is associated with aberrant pulmonary vascular remodeling that leads to increased pulmonary artery pressure, pulmonary vascular resistance, and right ventricular dysfunction. There is now accumulating evidence that the renin-angiotensin-aldosterone system is activated and contributes to cardiopulmonary remodeling that occurs in PAH. Increased plasma and lung tissue levels of angiotensin and aldosterone have been detected in experimental models of PAH and shown to correlate with cardiopulmonary hemodynamics and pulmonary vascular remodeling. These processes are abrogated by treatment with angiotensin receptor or mineralocorticoid receptor antagonists. At a cellular level, angiotensin and aldosterone activate oxidant stress signaling pathways that decrease levels of bioavailable nitric oxide, increase inflammation, and promote cell proliferation, migration, extracellular matrix remodeling, and fibrosis. Clinically, enhanced renin-angiotensin activity and elevated levels of aldosterone have been detected in patients with PAH, which suggests a role for angiotensin and mineralocorticoid receptor antagonists in the treatment of PAH. This review will examine the current evidence linking renin-angiotensin-aldosterone system activation to PAH with an emphasis on the cellular and molecular mechanisms that are modulated by aldosterone and may be of importance for the pathobiology of PAH.
Collapse
Affiliation(s)
- Bradley A. Maron
- Brigham and Women’s Hospital, Division of Cardiovascular Medicine, Boston, Massachusetts, USA
- Veterans Affairs Boston Healthcare System, Department of Cardiology, 1400 VFW Parkway, Boston, Massachusetts, USA
| | - Jane A. Leopold
- Brigham and Women’s Hospital, Division of Cardiovascular Medicine, Boston, Massachusetts, USA
| |
Collapse
|
28
|
The Renin-Angiotensin-aldosterone system in vascular inflammation and remodeling. Int J Inflam 2014; 2014:689360. [PMID: 24804145 PMCID: PMC3997861 DOI: 10.1155/2014/689360] [Citation(s) in RCA: 228] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/28/2014] [Accepted: 03/03/2014] [Indexed: 12/31/2022] Open
Abstract
The RAAS through its physiological effectors plays a key role in promoting and maintaining inflammation. Inflammation is an important mechanism in the development and progression of CVD such as hypertension and atherosclerosis. In addition to its main role in regulating blood pressure and its role in hypertension, RAAS has proinflammatory and profibrotic effects at cellular and molecular levels. Blocking RAAS provides beneficial effects for the treatment of cardiovascular and renal diseases. Evidence shows that inhibition of RAAS positively influences vascular remodeling thus improving CVD outcomes. The beneficial vascular effects of RAAS inhibition are likely due to decreasing vascular inflammation, oxidative stress, endothelial dysfunction, and positive effects on regeneration of endothelial progenitor cells. Inflammatory factors such as ICAM-1, VCAM-1, TNFα, IL-6, and CRP have key roles in mediating vascular inflammation and blocking RAAS negatively modulates the levels of these inflammatory molecules. Some of these inflammatory markers are clinically associated with CVD events. More studies are required to establish long-term effects of RAAS inhibition on vascular inflammation, vascular cells regeneration, and CVD clinical outcomes. This review presents important information on RAAS's role on vascular inflammation, vascular cells responses to RAAS, and inhibition of RAAS signaling in the context of vascular inflammation, vascular remodeling, and vascular inflammation-associated CVD. Nevertheless, the review also equates the need to rethink and rediscover new RAAS inhibitors.
Collapse
|
29
|
Iqbal J, Fay R, Adlam D, Squire I, Parviz Y, Gunn J, Pitt B, Zannad F. Effect of eplerenone in percutaneous coronary intervention-treated post-myocardial infarction patients with left ventricular systolic dysfunction: a subanalysis of the EPHESUS trial. Eur J Heart Fail 2014; 16:685-91. [DOI: 10.1002/ejhf.88] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/14/2014] [Accepted: 02/21/2014] [Indexed: 11/07/2022] Open
Affiliation(s)
- Javaid Iqbal
- Department of Cardiovascular Science at the University of Sheffield, and Cardiology Department at Sheffield Teaching Hospitals NHS Trust; Sheffield UK
| | - Renaud Fay
- INSERM, Centre d'Investigation Clinique and Centre Hospitalier Universitaire, and the Department of Cardiology; Nancy University, Université de Lorraine; Nancy France
| | - David Adlam
- Department of Cardiology, and NIHR Cardiovascular Biomedical Research Unit; Glenfield Hospital; Leicester UK
| | - Iain Squire
- Department of Cardiology, and NIHR Cardiovascular Biomedical Research Unit; Glenfield Hospital; Leicester UK
| | - Yasir Parviz
- Department of Cardiovascular Science at the University of Sheffield, and Cardiology Department at Sheffield Teaching Hospitals NHS Trust; Sheffield UK
| | - Julian Gunn
- Department of Cardiovascular Science at the University of Sheffield, and Cardiology Department at Sheffield Teaching Hospitals NHS Trust; Sheffield UK
| | - Bertram Pitt
- Cardiovascular Centre; University of Michigan; Ann Arbor MI USA
| | - Faiez Zannad
- INSERM, Centre d'Investigation Clinique and Centre Hospitalier Universitaire, and the Department of Cardiology; Nancy University, Université de Lorraine; Nancy France
| |
Collapse
|
30
|
Endothelial dysfunction in patients with primary aldosteronism: a biomarker of target organ damage. J Hum Hypertens 2014; 28:711-5. [PMID: 24553636 DOI: 10.1038/jhh.2014.11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 12/14/2013] [Accepted: 01/13/2014] [Indexed: 11/08/2022]
Abstract
Primary aldosteronism (PA) has been associated with increased target organ damage (TOD), most likely through mineralocorticoid receptor-dependent endothelial dysfunction, in comparison with essential hypertension (EH). The aim of this study was to evaluate the level of biomarkers of endothelial dysfunction in PA and the relationship with left ventricular hypertrophy (LVH) and microalbuminuria (MAU). A total of 50 PA patients and 51 patients with EH individually matched for age, sex, blood pressure and duration of hypertension participated in this study. Biomarkers of endothelial dysfunction, including von Willebrand factor (vWF), intercellular adhesion molecule 1 (ICAM-1) and oxidized low-density lipoprotein (ox-LDL), were measured. Plasma aldosterone concentration (PAC), MAU and echocardiography were also evaluated. In PA patients, vWF, ICAM-1, ox-LDL, LVH and MAU were all significantly higher than in EH patients (all P<0.05). Furthermore, LVH was positively correlated with PAC (P=0.002), vWF (P=0.013) and ox-LDL (P=0.020). MAU was positively correlated with PAC (P<0.001), vWF (P=0.013) and ICAM-1 (P=0.001). Multiple regression analysis indicated that vWF, ICAM-1 and PAC independently predicted MAU (all P<0.05). Likewise, PAC, vWF and ox-LDL were significant predictors of LVH (all P<0.05). Taken together, our results suggest that endothelial dysfunction may contribute to TOD in PA patients.
Collapse
|
31
|
Silvestre JS, Smadja DM, Lévy BI. Postischemic revascularization: from cellular and molecular mechanisms to clinical applications. Physiol Rev 2013; 93:1743-802. [PMID: 24137021 DOI: 10.1152/physrev.00006.2013] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the onset of ischemia, cardiac or skeletal muscle undergoes a continuum of molecular, cellular, and extracellular responses that determine the function and the remodeling of the ischemic tissue. Hypoxia-related pathways, immunoinflammatory balance, circulating or local vascular progenitor cells, as well as changes in hemodynamical forces within vascular wall trigger all the processes regulating vascular homeostasis, including vasculogenesis, angiogenesis, arteriogenesis, and collateral growth, which act in concert to establish a functional vascular network in ischemic zones. In patients with ischemic diseases, most of the cellular (mainly those involving bone marrow-derived cells and local stem/progenitor cells) and molecular mechanisms involved in the activation of vessel growth and vascular remodeling are markedly impaired by the deleterious microenvironment characterized by fibrosis, inflammation, hypoperfusion, and inhibition of endogenous angiogenic and regenerative programs. Furthermore, cardiovascular risk factors, including diabetes, hypercholesterolemia, hypertension, diabetes, and aging, constitute a deleterious macroenvironment that participates to the abrogation of postischemic revascularization and tissue regeneration observed in these patient populations. Thus stimulation of vessel growth and/or remodeling has emerged as a new therapeutic option in patients with ischemic diseases. Many strategies of therapeutic revascularization, based on the administration of growth factors or stem/progenitor cells from diverse sources, have been proposed and are currently tested in patients with peripheral arterial disease or cardiac diseases. This review provides an overview from our current knowledge regarding molecular and cellular mechanisms involved in postischemic revascularization, as well as advances in the clinical application of such strategies of therapeutic revascularization.
Collapse
|
32
|
Cubbon RM, Mercer BN, Sengupta A, Kearney MT. Importance of insulin resistance to vascular repair and regeneration. Free Radic Biol Med 2013; 60:246-63. [PMID: 23466555 DOI: 10.1016/j.freeradbiomed.2013.02.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 02/22/2013] [Accepted: 02/23/2013] [Indexed: 01/14/2023]
Abstract
Metabolic insulin resistance is apparent across a spectrum of clinical disorders, including obesity and diabetes, and is characterized by an adverse clustering of cardiovascular risk factors related to abnormal cellular responses to insulin. These disorders are becoming increasingly prevalent and represent a major global public health concern because of their association with significant increases in atherosclerosis-related mortality. Endogenous repair mechanisms are thought to retard the development of vascular disease, and a growing evidence base supports the adverse impact of the insulin-resistant phenotype upon indices of vascular repair. Beyond the impact of systemic metabolic changes, emerging data from murine studies also provide support for abnormal insulin signaling at the level of vascular cells in retarding vascular repair. Interrelated pathophysiological factors, including reduced nitric oxide bioavailability, oxidative stress, altered growth factor activity, and abnormal intracellular signaling, are likely to act in conjunction to impede vascular repair while also driving vascular damage. Understanding of these processes is shaping novel therapeutic paradigms that aim to promote vascular repair and regeneration, either by recruiting endogenous mechanisms or by the administration of cell-based therapies.
Collapse
Affiliation(s)
- Richard M Cubbon
- Multidisciplinary Cardiovascular Research Centre, LIGHT Laboratories, The University of Leeds, Leeds LS2 9JT, UK.
| | | | | | | |
Collapse
|
33
|
Zou HX, Jia J, Zhang WF, Sun ZJ, Zhao YF. Propranolol inhibits endothelial progenitor cell homing: a possible treatment mechanism of infantile hemangioma. Cardiovasc Pathol 2013; 22:203-10. [DOI: 10.1016/j.carpath.2012.10.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Revised: 10/04/2012] [Accepted: 10/05/2012] [Indexed: 12/17/2022] Open
|
34
|
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: 33] [Impact Index Per Article: 2.8] [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
| | - 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
| | | | - 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
|
35
|
He BJ, Anderson ME. Aldosterone and cardiovascular disease: the heart of the matter. Trends Endocrinol Metab 2013; 24:21-30. [PMID: 23040074 PMCID: PMC3532553 DOI: 10.1016/j.tem.2012.09.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Revised: 09/01/2012] [Accepted: 09/06/2012] [Indexed: 02/07/2023]
Abstract
Aldosterone contributes to the endocrine basis of heart failure, and studies on cardiac aldosterone signaling have reinforced its value as a therapeutic target. Recent focus has shifted to new roles of aldosterone that appear to depend on coexisting pathologic stimuli, cell type, and disease etiology. This review evaluates recent advances in mechanisms underlying aldosterone-induced cardiac disease and highlights the interplay between aldosterone and Ca(2+)/calmodulin dependent protein kinase II, whose hyperactivity during heart failure contributes to disease progression. Increasing evidence implicates aldosterone in diastolic dysfunction, and there is a need to develop more targeted therapeutics such as aldosterone synthase inhibitors and molecularly specific antioxidants. Despite accumulating knowledge, many questions still persist and will likely dictate areas of future research.
Collapse
Affiliation(s)
- B Julie He
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | |
Collapse
|
36
|
Zhao YH, Yuan B, Chen J, Feng DH, Zhao B, Qin C, Chen YF. Endothelial progenitor cells: therapeutic perspective for ischemic stroke. CNS Neurosci Ther 2012; 19:67-75. [PMID: 23230897 DOI: 10.1111/cns.12040] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 10/30/2012] [Accepted: 10/31/2012] [Indexed: 12/26/2022] Open
Abstract
Endothelial progenitor cells (EPCs), which can be cultured in vitro from mononuclear cells in peripheral blood or bone marrow, express both hematopoietic stem cell and endothelial cell markers on their surface. They are believed to participate in endothelial repair and postnatal angiogenesis due to their abilities of differentiating into endothelial cells and secreting protective cytokines and growth factors. Mounting evidence suggests that circulating EPCs are reduced and dysfunctional in various diseases including hypertension, diabetes, coronary heart disease, and ischemic stroke. Therefore, EPCs have been documented to be a potential biomarker for vascular diseases and a hopeful candidate for regenerative medicine. Ischemic stroke, as the major cause of disability and death, still has limited therapeutics based on the approaches of vascular recanalization or neuronal protection. Emerging evidence indicates that transplantation of EPCs is beneficial for the recovery of ischemic cerebral injury. EPC-based therapy could open a new avenue for ischemic cerebrovascular disease. Currently, clinical trials for evaluating EPC transfusion in treating ischemic stroke are underway. In this review, we summarize the general conceptions and the characteristics of EPCs, and highlight the recent research developments on EPCs. More importantly, the rationale, perspectives, and strategies for using them to treat ischemic stroke will be discussed.
Collapse
Affiliation(s)
- Yu-Hui Zhao
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Briet M, Schiffrin EL. Vascular actions of aldosterone. J Vasc Res 2012; 50:89-99. [PMID: 23172373 DOI: 10.1159/000345243] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 10/18/2012] [Indexed: 02/03/2023] Open
Abstract
Aldosterone exerts direct effects on the vascular system by inducing oxidative stress, inflammation, hypertrophic remodeling, fibrosis, and endothelial dysfunction. Aldosterone exerts its effects through genomic and nongenomic pathways in a mineralocorticoid receptor (MR)-dependent or independent manner. Other aldosterone receptors such as GPR30 have been identified. A tight relation exists between the aldosterone and angiotensin II pathways, as well as with the endothelin-1 system. There is a correlation between plasma levels of aldosterone and cardiovascular risk. Recently, an increasing body of evidence has underlined the importance of aldosterone in cardiovascular complications associated with the metabolic syndrome, such as arterial remodeling and endothelial dysfunction. Blockade of MR is an increasingly used evidence-based therapy for many forms of cardiovascular disease, including hypertension, heart failure, chronic kidney disease, and diabetes mellitus.
Collapse
Affiliation(s)
- Marie Briet
- Université Paris Descartes, Faculté de Medicine, Assistance Publique des Hôpitaux de Paris, Hôpital Européen Georges Pompidou, INSERM CIC 9201, Paris, France
| | | |
Collapse
|
38
|
Galuppo P, Bauersachs J. Mineralocorticoid receptor activation in myocardial infarction and failure: recent advances. Eur J Clin Invest 2012; 42:1112-20. [PMID: 22536780 DOI: 10.1111/j.1365-2362.2012.02676.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The classical view of aldosterone actions via the mineralocorticoid receptor (MR) limited to control of fluid balance and blood pressure homoeostasis has been progressively overcome by clinical and experimental evidence emphasizing the pleiotropic role of MR activation in the pathogenesis of cardiovascular disease. Clinical studies have shown the benefit of MR blockade in patients with left ventricular dysfunction and heart failure after myocardial infarction (MI), hypertension or diabetic nephropathy. Deleterious effects of MR activation include cardiac structural and electrical remodelling, cardiovascular fibrosis, inflammation and oxidative stress. Complexity of pathophysiological role of MR derives from the presence of circulating glucocorticoids at higher concentrations than aldosterone and the equal affinity of the MR for aldosterone, cortisol and corticosterone. Recent experimental studies using different animal models and genetic tools have deeply explored the cell-specific functional role of MR in cardiovascular pathology. This review addresses emerging preclinical studies as well as ongoing clinical trials regarding MR activation in MI and failure.
Collapse
Affiliation(s)
- Paolo Galuppo
- Klinik fuer Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | | |
Collapse
|
39
|
Jung C, Florvaag A, Oberle V, Fritzenwanger M, Kretschmar D, Kuethe F, Betge S, Goebel B, Franz M, Barz D, Ferrari M, Figulla HR. Positive effect of eplerenone treatment on endothelial progenitor cells in patients with chronic heart failure. J Renin Angiotensin Aldosterone Syst 2012; 13:401-6. [DOI: 10.1177/1470320312447650] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background: Endothelial progenitor cells (EPCs) are known to play a significant role in reendothelialization and vascular repair. Recently, a mineralocorticoid receptor was demonstrated to be expressed by EPCs. The study aimed to evaluate a potential influence of eplerenone treatment on the total number of EPCs in patients with chronic heart failure. Methods: Eighty-seven male patients with chronic heart failure were included (age: 23–83 years; body mass index 29.1 ± 5.1 kg/m2; New York Heart failure classification (NYHA) I: 29 patients, NYHA II: 32 patients, NYHA III: 26 patients). Numbers of circulating EPCs were quantified immediately using flow cytometry. Twenty-eight patients received therapy with eplerenone. Patients were further characterized by echocardiography, spirometry and laboratory markers. Results: Patients with ongoing eplerenone administration showed higher levels of circulating cells expressing CD34+ ( p<0.05) and CD34+KDR+ ( p<0.05) and CD34+CD133+KDR+ cells ( p<0.05). The effects of eplerenone treatment could be shown to be independent of NYHA status, genesis of the underlying cardiovascular morbidity, left ventricular function and co-medication. Conclusion: Patients with chronic heart failure treated with eplerenone show higher numbers of EPCs. The clinical benefit for treatment with eplerenone has been demonstrated even for patients with mild heart failure and might be partially mediated by EPCs.
Collapse
Affiliation(s)
- Christian Jung
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Anna Florvaag
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Volker Oberle
- Division of Cardiology, Institute for Transfusion Medicine, Friedrich-Schiller-University Jena, Germany
| | | | - Daniel Kretschmar
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Friedhelm Kuethe
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Stefan Betge
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Bjoern Goebel
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Marcus Franz
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Dagmar Barz
- Division of Cardiology, Institute for Transfusion Medicine, Friedrich-Schiller-University Jena, Germany
| | - Markus Ferrari
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Hans R Figulla
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| |
Collapse
|
40
|
Van Laecke S, Nagler EVT, Vanholder R. Thrombotic microangiopathy: a role for magnesium? Thromb Haemost 2012; 107:399-408. [PMID: 22274299 DOI: 10.1160/th11-08-0593] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Accepted: 12/01/2011] [Indexed: 12/15/2022]
Abstract
Despite advances in more recent years, the pathophysiology and especially treatment modalities of thrombotic microangiopathy (TMA) largely remain enigmatic. Disruption of endothelial homeostasis plays an essential role in TMA. Considering the proven causal association between magnesium and both endothelial function and platelet aggregability, we speculate that a magnesium deficit could influence the course of TMA and the related haemolytic uraemic syndrome and thrombotic thrombocytopenic purpura. A predisposition towards TMA is seen in many conditions with both extracellular and intracellular magnesium deficiency. We propose a rationale for magnesium supplementation in TMA, in analogy with its evidence-based therapeutic application in pre-eclampsia and suggest, based on theoretical grounds, that it might attenuate the development of TMA, minimise its severity and prevent its recurrence. This is based on several lines of evidence from both in vitro and in vivo data showing dose-dependent effects of magnesium supplementation on nitric oxide production, platelet aggregability and inflammation. Our hypothesis, which is further amenable to assessment in animal models before therapeutic applications in humans are implemented, could be explored both in vitro and in vivo to decipher the potential role of magnesium deficit in TMA and of the effects of its supplementation.
Collapse
Affiliation(s)
- Steven Van Laecke
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium.
| | | | | |
Collapse
|
41
|
Gustafsson F, Azizi M, Bauersachs J, Jaisser F, Rossignol P. Targeting the aldosterone pathway in cardiovascular disease. Fundam Clin Pharmacol 2011; 26:135-45. [PMID: 22044496 DOI: 10.1111/j.1472-8206.2011.01004.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Accumulated evidence has demonstrated that aldosterone is a key player in the pathogenesis of cardiovascular (CV) disease. Multiple clinical trials have documented that intervention in the aldosterone pathway can reduce blood pressure and lower albuminuria and improve outcome in patients with heart failure or myocardial infarction. Recent studies have unraveled details about the role of aldosterone at the cellular level in CV disease. The relative importance of glucocorticoids and aldosterone in terms of mineralocorticoid receptor activation is currently being debated. Also, studies are addressing which aldosterone modulator to use, which timing of treatment to aim for, and in which population to intervene. This review provides an overview of recent developments in the understanding of the role of aldosterone in CV disease, with particular reference to mechanisms and potential targets of intervention. Finally, ongoing or desirable clinical trials in the field are highlighted. The review is partly based on discussions between basic scientists and clinical trialists at the Cardiovascular Clinical Trials Forum 2009 and subsequently updated to encompass the most recent developments.
Collapse
Affiliation(s)
- Finn Gustafsson
- Department of Cardiology, Rigshospitalet, Copenhagen, Denmark.
| | | | | | | | | |
Collapse
|
42
|
Wu VC, Lo SC, Chen YL, Huang PH, Tsai CT, Liang CJ, Kuo CC, Kuo YS, Lee BC, Wu EL, Lin YH, Sun YY, Lin SL, Chen JW, Lin SJ, Wu KD. Endothelial progenitor cells in primary aldosteronism: a biomarker of severity for aldosterone vasculopathy and prognosis. J Clin Endocrinol Metab 2011; 96:3175-83. [PMID: 21816781 DOI: 10.1210/jc.2011-1135] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
CONTEXT Primary aldosteronism (PA) is associated with a higher incidence of cardiovascular events, probably through mineralocorticoid receptor (MR)-dependent endothelial cell dysfunction, in comparison with essential hypertension (EH). OBJECTIVE Our objective was to investigate the number and function of endothelial progenitor cells (EPC) in PA and the relationship with arterial stiffness and disease progression. DESIGN AND SETTING We conducted a prospective study of the change of EPC number and outcome of PA patients after treatment at a tertiary medical center. PRIMARY OUTCOMES Changes in arterial stiffness and EPC number after treatment and the curability of hypertension were assessed. PATIENTS A total of 113 PA patients (87 patients diagnosed with aldosterone-producing adenoma, 26 with idiopathic hyperaldosteronism) and 55 patients with EH participated. RESULTS PA patients had higher arterial stiffness than EH patients (P = 0.006), with a lower numbers of circulating EPC and endothelial colony-forming units (P < 0.05). The differences were ameliorated at 6 months after unilateral adrenalectomy or treatment with spironolactone. Expression of MR was identified in the EPC. The number of circulating EPC was inversely correlated with the plasma aldosterone concentration (P = 0.021), arterial stiffness (P = 0.029) and serum high-sensitivity C-reactive protein (P = 0.03). High-dose aldosterone (10(-5) and 10(-6) m) attenuated EPC proliferation and angiogenesis in vitro. Among the 45 patients who underwent unilateral adrenalectomy, 32 (71%) were cured of hypertension. The preoperative number of EPC [log(EPC number percent) >-3.6] predicted the curability of hypertension after adrenalectomy (P = 0.003). CONCLUSIONS The relative deficiency of EPC in PA patients may contribute to aldosterone vasculopathy, which can be reversed by adrenalectomy and spironolactone. High aldosterone levels attenuated EPC proliferation and angiogenesis. Circulating EPC number may be a valuable biomarker to identify PA patients with a high incidence of arterial stiffness and to predict postoperative residual hypertension of aldosterone-producing adenoma.
Collapse
Affiliation(s)
- Vin-Cent Wu
- Department of Internal Medicine, Taipei Veterans General Hospital and Cardiovascular Research Center, National Yang-Ming University, Taipei 100, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
MicroRNAs (miRNAs) are important regulators of gene expression and fundamentally impact on cardiovascular function in health and disease. A tight control of miRNA expression is crucial for the maintenance of tissue homeostasis. However, a comprehensive understanding of the various levels of miRNA regulation is in its infancy. We here summarize the current knowledge about regulation of cardiovascular miRNAs at the transcriptional level by transcription factors, during processing by the Drosha and Dicer complexes and the importance of miRNA modification, editing, and decay mechanisms. As an example, miRNA regulation in diabetic and hypoxic cardiovascular disease conditions is discussed. Better knowledge about regulatory mechanisms of miRNAs in cardiovascular disease will probably lead to improved and novel miRNA-based therapeutic therapies.
Collapse
Affiliation(s)
- Johann Bauersachs
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Carl-Neuberg-Strasse 1, Hannover, Germany.
| | | |
Collapse
|
44
|
Boffa JJ, Chauvet S, Mihout F. [Slowing chronic kidney disease progression: hopes and disappointments. Vascular repair of chronic kidney]. Presse Med 2011; 40:1065-73. [PMID: 21889290 DOI: 10.1016/j.lpm.2011.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 06/07/2011] [Accepted: 06/10/2011] [Indexed: 10/17/2022] Open
Abstract
In chronic kidney disease patients, inexorable renal function decline is reduced by renin-angiotensin system (RAS) blockers. ACE inhibitors and angiotensin receptor blockers decrease blood pressure and proteinuria. Guidelines recommend a reduction of blood pressure to less than 130/80 mmHg and urinary protein excretion below 0.5 g/d. The combined use of a diuretic increases anti-proteinuric effect and blood pressure control of RAS blockers. Drugs as mineralo-corticocoids receptor antagonist and endothelin receptor antagonists reduce further albuminuria in combination with RAS blocker, but side effects need to be precised. Both metabolic acidosis and hyperuricemia represent new therapeutic goals to slow renal function decline in CKD patients. Renal fibrosis treatment and regenerative medicine are stemming and will be important issues for kidney and other organs in the future.
Collapse
|
45
|
Fleissner F, Thum T. Critical role of the nitric oxide/reactive oxygen species balance in endothelial progenitor dysfunction. Antioxid Redox Signal 2011; 15:933-48. [PMID: 20712407 PMCID: PMC3135185 DOI: 10.1089/ars.2010.3502] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 08/13/2010] [Indexed: 12/12/2022]
Abstract
Endothelial injury and dysfunction are critical events in the pathogenesis of cardiovascular disease. During these processes, an impaired balance of nitric oxide bioavailability and oxidative stress is mechanistically involved. Circulating angiogenic cells (including early and late outgrowth endothelial progenitor cells (EPC)) contribute to formation of new blood vessels, neovascularization, and homeostasis of the vasculature, and are highly sensitive for misbalance between NO and oxidative stress. We here review the role of the endothelial nitric oxide synthase and oxidative stress producing enzyme systems in EPC during cardiovascular disease. We also focus on the underlying molecular mechanisms and potential emerging drug- and gene-based therapeutic strategies to improve EPC function in cardiovascular diseased patients.
Collapse
Affiliation(s)
- Felix Fleissner
- Institute of Molecular and Translational Therapeutic Strategies, IFB-Tx, Hannover Medical School, Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, IFB-Tx, Hannover Medical School, Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
46
|
Becher UM, Endtmann C, Tiyerili V, Nickenig G, Werner N. Endothelial damage and regeneration: the role of the renin-angiotensin-aldosterone system. Curr Hypertens Rep 2011; 13:86-92. [PMID: 21108024 DOI: 10.1007/s11906-010-0171-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The renin-angiotensin-aldosterone system (RAAS) is part of the blood pressure regulating system. Its main effector peptide is angiotensin II (Ang II). Although it may induce hypertension, the proinflammatory, profibrotic, and prothrombotic effects are mainly mediated by effects of Ang II on the cellular and molecular level that are independent of blood pressure. Therefore, pharmacotherapeutic intervention within the RAAS is an important treatment modality for patients suffering from cardiovascular diseases, even those who are not hypertensive. In addition to the blood pressure lowering and vasculoprotective (pleiotropic) effects of angiotensin II type 1 (AT(1)) receptor blockers (ARBs), and angiotensin-converting enzyme (ACE) inhibitors, regenerative progenitor cell therapy emerges as an auxiliary therapy to improve regeneration of the vascular endothelium. This review focuses on the growing knowledge about regenerating vascular cells, their response to RAAS effectors, and RAAS-modulating pharmacotherapy in the context of endothelial cell damage and regeneration.
Collapse
Affiliation(s)
- Ulrich M Becher
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Sigmund-Freud-Strasse 25, Bonn, Germany.
| | | | | | | | | |
Collapse
|