51
|
He YP, Zhao LY, Zheng QS, Liu SW, Zhao XY, Lu XL, Niu XL, Li X. Involvement of ERK and AKT signaling in the growth effect of arginine vasopressin on adult rat cardiac fibroblast and the modulation by simvastatin. Mol Cell Biochem 2008; 317:33-41. [DOI: 10.1007/s11010-008-9802-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Accepted: 05/23/2008] [Indexed: 10/21/2022]
|
52
|
Liu J, Shen Q, Wu Y. Simvastatin prevents cardiac hypertrophy in vitro and in vivo via JAK/STAT pathway. Life Sci 2008; 82:991-6. [DOI: 10.1016/j.lfs.2008.02.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Revised: 01/23/2008] [Accepted: 02/10/2008] [Indexed: 10/22/2022]
|
53
|
Atorvastatin Attenuates Cardiomyocyte Loss in Adult Rats From Protein-Restricted Dams. J Card Fail 2008; 14:151-60. [DOI: 10.1016/j.cardfail.2007.10.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2007] [Revised: 10/04/2007] [Accepted: 10/12/2007] [Indexed: 01/09/2023]
|
54
|
Kato N, Liang YQ, Ochiai Y, Jesmin S. Systemic evaluation of gene expression changes in major target organs induced by atorvastatin. Eur J Pharmacol 2008; 584:376-89. [PMID: 18295756 DOI: 10.1016/j.ejphar.2008.01.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Revised: 12/25/2007] [Accepted: 01/21/2008] [Indexed: 11/15/2022]
Abstract
Statins have been reported to protect against end-organ damage in essential hypertension; however, detailed mechanisms underlying organ-protective actions of statins remain unclear. Statins can exert pleiotropic effects aside from lowering cholesterol and blood pressure levels through several different pathways, which may lead to distinct patterns of changes in gene expression in vital end-organs. The aim of the present study was to systemically evaluate gene expression changes in three major end-organs (the brain, heart and kidney) induced by atorvastatin at a dose that altered neither blood pressure nor plasma total cholesterol levels. The stroke-prone spontaneously hypertensive (SHRSP) rats, an established model of hypertension and end-organ damage, was treated with atorvastatin (15 mg/kg/day) for 4 weeks from 12 to 16 weeks of age. DNA microarray technology was used to identify gene expression changes in three end-organs. In the current experimental setting, 4 weeks of atorvastatin treatment lowered plasma levels of non-esterified fatty acid significantly (P=0.0012) and triglyceride modestly (P=0.07) without altering blood pressure and plasma total cholesterol levels in male SHRSP rats. The level of expression of a number of genes was changed in an organ-specific manner after 4 weeks of drug administration to SHRSP rats. Among the end-organs studied, the most prominent alteration in gene expression was observed in the heart. The identical treatment protocol was applied to age-matched normotensive control rats, Wistar Kyoto rats, and this also caused a number of genes to be differentially expressed in an organ-specific manner. These results provide new insights into the mechanisms underlying the potential efficacy of statins in protecting against end-organ damage in essential hypertension and thus lay the foundation for future studies.
Collapse
Affiliation(s)
- Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, International Medical Center of Japan, Tokyo, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan.
| | | | | | | |
Collapse
|
55
|
Horwich TB, MacLellan WR. Atorvastatin and statins in the treatment of heart failure. Expert Opin Pharmacother 2007; 8:3061-8. [DOI: 10.1517/14656566.8.17.3061] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
56
|
Abstract
Left ventricular hypertrophy (LVH), despite its adaptive nature, is associated with an increased risk of cardiovascular morbidity and mortality. Achievement of LVH regression is thus considered a principal therapeutic aim. However, regression of LVH induced by various therapeutic means may exhibit differing patterns, with variable biological implications. Inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase (statins) have been shown to induce prevention or regression of LVH in different models of pathological myocardial growth. In addition to reduction of LV mass, statins were shown to reduce myocardial fibrosis, increase capillary density network and attenuate electrical instability of the hypertrophied heart. Most importantly, statins improved systolic and diastolic LV function and even decreased mortality. The inhibition of hypertrophic growth was only partly achieved by reduction of haemodynamic overload. Direct mechanisms, such as inhibition of neurohumoral activation in the myocardial tissue, attenuated production of growth factors and markers of inflammation and reduction of oxidative stress also seem to participate. The protective effect of statins was associated with the inhibition of expression and activation of small guanosintriphosphate-binding proteins such as Ras and Rho, which control the intensity of oxidative stress, the production and availability of nitric oxide, and the expression of genes involved in myocardial growth. In addition to reduction of LV mass, statins may also improve the prognosis of LVH independently of their lipid-lowering effect.
Collapse
Affiliation(s)
- F Simko
- School of Medicine, Komensky University, Bratislava, Slovak Republic.
| |
Collapse
|
57
|
McKinsey TA, Kass DA. Small-molecule therapies for cardiac hypertrophy: moving beneath the cell surface. Nat Rev Drug Discov 2007; 6:617-35. [PMID: 17643091 DOI: 10.1038/nrd2193] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pathological stress from cardiovascular disease stimulates hypertrophy of heart cells, which increases the risk of cardiac morbidity and mortality. Recent evidence has indicated that inhibiting such hypertrophy could be beneficial, encouraging drug discovery and development efforts for agents that could achieve this goal. Most existing therapies that have antihypertrophic effects target outside-in signalling in cardiac cells, but their effectiveness seems limited, and so attention has recently turned to the potential of targeting intracellular signalling pathways. Here, we focus on new developments with small-molecule inhibitors of cardiac hypertrophy, summarizing both agents that have been in or are poised for clinical testing, and pathways that offer further promising potential therapeutic targets.
Collapse
Affiliation(s)
- Timothy A McKinsey
- Gilead Colorado, Inc., 7575 West 103rd Avenue, Westminster, Colorado 80021, USA.
| | | |
Collapse
|
58
|
Habibi J, Whaley-Connell A, Qazi MA, Hayden MR, Cooper SA, Tramontano A, Thyfault J, Stump C, Ferrario C, Muniyappa R, Sowers JR. Rosuvastatin, a 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitor, decreases cardiac oxidative stress and remodeling in Ren2 transgenic rats. Endocrinology 2007; 148:2181-8. [PMID: 17317778 DOI: 10.1210/en.2006-1355] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Angiotensin-II (Ang-II)-stimulated increases in nicotinamide adenine dinucleotide phosphate reduced (NADPH) oxidase activity and oxidative stress are known to play a key role in cardiac remodeling. Inhibition of isoprenylation and activation of small G proteins, such as Rac1, a component of NADPH oxidase, may mediate the antioxidant actions of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins). In this study, we investigated the effects of rosuvastatin on cardiac oxidative stress and remodeling in transgenic rats (Ren2) overexpressing the mouse renin gene with elevated cardiac levels of Ang-II. We treated 6- to 7-wk-old Ren2 rats and age-matched Sprague-Dawley (SD) rats with rosuvastatin (10 mg/kg.d) or vehicle for 3 wk. At the end of the treatment period, left ventricular mass, wall thickness, ejection fraction (by echocardiography), and cardiac remodeling (by light microscopy and immunohistochemistry) were assessed. In addition, myocardial content of nitrotyrosine, malondialdehyde, NADPH-oxidase subunits (gp91(phox), p40(phox), and p22(phox)), and Rac1 were analyzed by immunochemistry. Systolic blood pressure was significantly higher in Ren2 rats, compared with SD rats (P < 0.05); rosuvastatin had no significant effect on systolic blood pressure in either group. In Ren2, but not SD rats, rosuvastatin significantly improved the ventricular ejection fraction, cardiac hypertrophy, and perivascular fibrosis (P < 0.05). In addition, rosuvastatin administration significantly decreased the accentuated myocardial gp91(phox), p40(phox), p22(phox), and Rac1 expression. These changes were accompanied by a parallel reduction in myocardial lipid peroxidation (nitrotyrosine and malondialdehyde content) (P < 0.05). These results suggest that in vivo statin treatment through its direct actions on the heart reduces oxidative stress and remodeling including ventricular mass regression in the Ang-II-dependent Ren2 model.
Collapse
Affiliation(s)
- Javad Habibi
- University of Missouri School of Medicine, Columbia, Missouri 65212, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Huan Loh P, Windram JD, Tin L, Reddy P, Velavan P, Rigby AS, Atkin P, Nikitin NP, Clark AL, Cleland JGF. The effects of initiation or continuation of statin therapy on cholesterol level and all-cause mortality after the diagnosis of left ventricular systolic dysfunction. Am Heart J 2007; 153:537-44. [PMID: 17383290 DOI: 10.1016/j.ahj.2007.01.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Accepted: 01/22/2007] [Indexed: 11/18/2022]
Abstract
BACKGROUND Statins (3-hydroxy-3-methyl-glutaryl coenzyme A reductase inhibitors) are known to reduce mortality and cardiac events in patients with coronary artery disease who have not progressed to left ventricular systolic dysfunction (LVSD) and/or heart failure (HF). This study investigated the effect of changes in statin therapy and cholesterol level on mortality in patients with LVSD. METHODS Data from consecutive patients with LVSD enrolled in a single local hospital HF management program were analyzed. Patients were grouped according to changes in statin treatment within 4 months after their initial visit: groups NS (no statin), IS (initiation of statin), CS (continuation of statin), and SS (statin stopped). RESULTS Nine hundred patients were followed for a median of 36 (28-43) months (range, 16-66 months). The 2-year mortality was 16.7%. Groups IS and CS had lower 2-year mortality than groups NS and SS (11.0% and 11.9% vs 22.0% and 34.8%, respectively; P < .001). This was independent of age, sex, severity of LVSD, HF medications, New York Heart Association functional class, and baseline cholesterol. The effect was mainly observed in patients with coronary artery disease. In 734 patients who had completed 1-year follow-up on stable HF treatment, neither baseline cholesterol nor change over 1 year predicted outcome. CONCLUSION Initiation and maintenance of treatment with statins is associated with better survival in patients with LVSD. This could not be explained by other measured variables.
Collapse
Affiliation(s)
- Poay Huan Loh
- Academic Cardiology Department, University of Hull, Hull Royal Infirmary, Hull, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Krum H, Ashton E, Reid C, Kalff V, Rogers J, Amarena J, Singh B, Tonkin A. Double-Blind, Randomized, Placebo-Controlled Study of High-Dose HMG CoA Reductase Inhibitor Therapy on Ventricular Remodeling, Pro-Inflammatory Cytokines and Neurohormonal Parameters in Patients With Chronic Systolic Heart Failure. J Card Fail 2007; 13:1-7. [PMID: 17338996 DOI: 10.1016/j.cardfail.2006.09.008] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2006] [Revised: 09/18/2006] [Accepted: 09/28/2006] [Indexed: 11/16/2022]
Abstract
BACKGROUND Statins decrease mortality in patients with coronary artery disease. However, chronic heart failure (CHF) patients were often excluded in such trials. Statins possess pharmacologic properties (independent of cholesterol lowering) that may be beneficial on ventricular remodeling in such patients. METHODS AND RESULTS We conducted a 6-month randomized placebo (PBO)-controlled study of rosuvastatin (ROS) in patients with systolic (left ventricular ejection fraction [LVEF] <40%) CHF of ischemic or nonischemic etiology. The primary end point was change in LVEF by radionuclide ventriculogram. Secondary end points included change in echocardiographic parameters, neurohormonal and inflammatory markers, Packer composite score, death, and heart failure hospitalization. Patients were well matched for baseline values. Compared with PBO (n = 46), ROS patients (n = 40) had a decrease in low-density lipoprotein cholesterol (PBO +3, ROS -54%, P < .001). There was no significant change in LVEF by radionuclide ventriculogram (PBO +5.3, ROS +3.2%), fractional shortening by echocardiographic (PBO +2.7, ROS +1.8%), left ventricular end-diastolic diameter (PBO -1.7, ROS +0.8 mm), left ventricular end-systolic diameter (PBO -1.9, ROS +0.1 mm). Plasma norepinephrine, endothelin-1, brain natriuretic peptide, hsCRP, tumor necrosis factor-alpha and interleukin-6, patient global assessment, Packer composite, death/heart failure hospitalization, and adverse events were similar between PBO and ROS. CONCLUSIONS Despite being safe and effective at decreasing plasma cholesterol, high-dose ROS did not beneficially alter parameters of LV remodeling. Reasons for absence of benefit are uncertain, but may include patient population studied, high dose of ROS used or high use of effective background CHF medications.
Collapse
Affiliation(s)
- Henry Krum
- Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Tamargo J, Caballero R, Gómez R, Núñez L, Vaquero M, Delpón E. Lipid-lowering therapy with statins, a new approach to antiarrhythmic therapy. Pharmacol Ther 2006; 114:107-26. [PMID: 17287023 DOI: 10.1016/j.pharmthera.2006.12.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2006] [Accepted: 12/08/2006] [Indexed: 11/17/2022]
Abstract
Inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase (statins) are the most effective and best-tolerated drugs to treat elevated levels of low-density lipoprotein cholesterol (LDL-C). In addition, they exhibit other effects unrelated to their lipid lowering effects (pleiotropic actions). In recent years, experimental and clinical evidence demonstrates that statins exert antiarrhythmic properties, reducing the recurrences of supraventricular and life-threatening ventricular arrhythmias both in patients with and without coronary artery disease (CAD). Thus, statins may constitute a novel therapeutic approach to cardiac arrhythmias. This article reviews the antiarrhythmic properties of statins as well as the possible mechanisms involved, including the lowering of LDL-C levels, the improvement of endothelial dysfunction and autonomic function, the stabilization of the atherosclerotic plaques, the antioxidant, antiinflammatory, antithrombotic and cardioprotective properties and the modulation of transmembrane ion fluxes.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
62
|
Frick M, Alber HF, Hügel H, Schwarzacher SP, Pachinger O, Weidinger F. Short- and long-term changes of flow-mediated vasodilation in patients under statin therapy. Clin Cardiol 2006; 25:291-4. [PMID: 12058793 PMCID: PMC6654712 DOI: 10.1002/clc.4960250610] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Flow-mediated vasodilation (FMD) of the brachial artery (BA) has been shown to improve in response to lipid-lowering therapy and other therapeutic interventions, usually within 1 to 2 months. Whether FMD remains improved under therapy in the longer term is unknown. HYPOTHESIS The aim of this study was to examine the short- and long-term changes of FMD under statin therapy. METHODS Flow-mediated vasodilation and nitroglycerin-mediated vasodilation (NMD) of the BA were measured with high-resolution ultrasound (13 MHz) at baseline and at 4 and 10 months in 18 consecutively recruited patients with coronary artery disease (CAD), in whom statin therapy was newly established. RESULTS The decrease of total plasma cholesterol levels after 4 and 10 months of statin therapy (243 +/- 31 vs. 186 +/- 30 vs. 191 +/- 40 mg/dl; p < 0.001) was accompanied by an increase in FMD from 4.4 +/- 3.8% at baseline to 9.6 +/- 2.7% at 4 months and to 9.5 +/- 2.6% at 10 months (p < 0.001). Nitroglycerin-mediated vasodilation showed a trend toward improvement after 4 months (14.6 +/- 7.5 vs. 19.1 +/- 3.6 vs. 19.4 +/- 5.6%; NS). The FMD/NMD ratio also rose significantly after 4 months and remained improved after 10 months of statin therapy (0.31 +/- 0.25 vs. 0.52 +/- 0.16 vs. 0.50 +/- 0.14; p < 0.01). CONCLUSION Statin therapy is associated with sustained improvement of endothelial function up to 10 months. These data support the utility of FMD for the assessment of vascular function in response to lipid-lowering therapy or other therapeutic interventions in long-term studies.
Collapse
Affiliation(s)
- Matthias Frick
- Division of Cardiology, Department of Medicine, University of Innsbruck, Innsbruck, Austria
| | - Hannes F. Alber
- Division of Cardiology, Department of Medicine, University of Innsbruck, Innsbruck, Austria
| | - Heike Hügel
- Division of Cardiology, Department of Medicine, University of Innsbruck, Innsbruck, Austria
| | | | - Otmar Pachinger
- Division of Cardiology, Department of Medicine, University of Innsbruck, Innsbruck, Austria
| | - Franz Weidinger
- Division of Cardiology, Department of Medicine, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
63
|
Kurian KC, Rai P, Sankaran S, Jacob B, Chiong J, Miller AB. The effect of statins in heart failure: beyond its cholesterol-lowering effect. J Card Fail 2006; 12:473-8. [PMID: 16911915 DOI: 10.1016/j.cardfail.2006.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Revised: 01/29/2006] [Accepted: 02/03/2006] [Indexed: 10/24/2022]
Abstract
BACKGROUND Statins known as 3-hydroxyl-3-methyl-glutaryl-coenzyme A (HMG-CoA) are designed to lower plasma cholesterol levels. They are used to treat hypercholesterolemia, ischemic heart disease patients, heart transplant recipients, in prevention of Alzheimer's dementia, multiple sclerosis, and have also been shown to reduce cancer risk. METHODS AND RESULTS The idea of statin treatment in chronic heart failure is not well established. It has been shown to be beneficial in patients with ischemic heart disease with heart failure. Emerging trends show their usefulness in patients with nonischemic heart failure. Statins exhibit pleiotropic effects in stabilizing the atherosclerotic plaques, improvement of endothelial function, inhibition of cell migration and proliferation, and reduction of inflammation and oxidative stress. They also improve autonomic function with an increased parasympathetic drive, downregulate the angiotensin II type I receptors, and induce angiogenesis. CONCLUSION This article is a review on the current knowledge on statin use in heart failure.
Collapse
Affiliation(s)
- Kizhake C Kurian
- Division of Cardiology, University of Florida Health Science Center-Jacksonville, Jacksonville, Florida 32223, USA
| | | | | | | | | | | |
Collapse
|
64
|
Lecian D, Demova H, Lodererova A, Zdychova J, Kluckova H, Teplan V, Voska L, Komers R. Renal Effects of HMG-CoA Reductase Inhibition in a Rat Model of Chronic Inhibition of Nitric Oxide Synthesis. Kidney Blood Press Res 2006; 29:135-43. [PMID: 16899993 DOI: 10.1159/000094988] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2005] [Accepted: 04/11/2006] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS In addition to their lipid-lowering effects, HMG-CoA reductase inhibitors (statins) induce a variety of pleiotropic actions that have been recently studied in the area of cardiovascular and renal protection. In the present studies we sought to determine whether statins retain beneficial effects in the experimental model of NO deficiency achieved by chronic administration of a pressor dose of L-arginine analogue N-nitro-L-arginine-methyl ester (L-NAME). METHODS To address this issue, blood pressure (BP), renal function (GFR), and albuminuria were determined in rats treated for 4 weeks with L-NAME, L-NAME + atorvastatin (ATO), and in untreated controls. In addition, renal cortical protein expression of caveolin 1 (CAV1), vascular endothelial growth factor (VEGF), and activity of RhoA were also determined. RESULTS L-NAME administration resulted in sustained elevation of BP, decreased GFR, and in higher albuminuria as compared to control animals. Co-administration of ATO with L-NAME normalized albuminuria and prevented decreases in GFR in L-NAME rats without having an impact on pressor effects of L-NAME. CAV1 protein expression was similar in all groups of rats. In contrast, VEGF expression and RhoA activity was increased in L-NAME-treated animals, and normalized with co-administration of ATO. CONCLUSION Treatment with ATO exerts early nephroprotective effects in the NO-deficient model of hypertension. These effects could be mediated by amelioration of VEGF expression and reduction of RhoA activity.
Collapse
Affiliation(s)
- Dalibor Lecian
- Department of Nephrology, Institute for Clinical and Experimental Medicine, Charles University, Prague, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
65
|
Khush KK, Waters DD. Effects of Statin Therapy on the Development and Progression of Heart Failure: Mechanisms and Clinical Trials. J Card Fail 2006; 12:664-74. [PMID: 17045188 DOI: 10.1016/j.cardfail.2006.05.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Revised: 04/28/2006] [Accepted: 05/01/2006] [Indexed: 11/18/2022]
Abstract
BACKGROUND Statin therapy has been shown to effectively lower low-density lipoprotein cholesterol levels and reduce cardiovascular events. Statins also appear to exert other favorable effects, including anti-inflammatory actions and improvement in endothelial function. Statin therapy may therefore yield important clinical benefits in patients with heart failure-a physiologic state characterized by systemic inflammation and endothelial dysfunction. METHODS AND RESULTS This review summarizes basic and clinical investigations regarding the role of statin therapy in heart failure, focusing on potential mechanisms and preliminary clinical data. There is now extensive evidence suggesting that statins improve endothelial function, inhibit neurohormonal activation, restore autonomic balance, reduce inflammation, and prevent ventricular remodeling. Retrospective and small-scale prospective studies suggest that statins prevent the development of heart failure and reduce mortality in patients with established HF. CONCLUSION Preliminary evidence supports a role for statins in improving surrogate markers and clinical outcomes in ischemic and nonischemic heart failure. Large-scale randomized clinical trials are needed to definitively address this important topic.
Collapse
Affiliation(s)
- Kiran K Khush
- Division of Cardiology, University of California, San Francisco School of Medicine, San Francisco, California 94143, USA
| | | |
Collapse
|
66
|
Krum H, Bailey M, Meyer W, Verkenne P, Dargie H, Lechat P, Anker S. Impact of Statin Therapy on Clinical Outcomes in Chronic Heart Failure Patients According to Beta-Blocker Use: Results of CIBIS II. Cardiology 2006; 108:28-34. [PMID: 16960445 DOI: 10.1159/000095629] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Accepted: 05/22/2006] [Indexed: 12/13/2022]
Abstract
BACKGROUND HMG-CoA reductase inhibitors (statins) are widely prescribed in patients with established systolic chronic heart failure (CHF). However, there is considerable controversy regarding their benefit in this setting. We therefore conducted a post-hoc analysis of outcomes according to statin use within the Second Cardiac Insufficiency Bisoprolol Study of the beta-blocker, bisoprolol, in NYHA classes III-IV systolic CHF patients (left ventricular ejection fraction <35%), receiving background ACE inhibitor and diuretics. METHODS Analysis of clinical outcomes was performed according to baseline use of statins and subsequent randomisation to placebo or bisoprolol. Cumulative incidence curves for clinical events were constructed using the Kaplan-Meier method and tested for significance by log-rank statistic. Multivariate analysis was performed using the Cox proportional hazards regression model. RESULTS Two hundred and twenty-six of 2,647 patients were receiving statins at baseline (8.5%). Patients were well-matched in the 4 study groups at baseline for gender, weight, NYHA class and LVEF, however statin/bisoprolol patients were significantly younger (p < 0.05). Statin use at baseline was associated with a significant survival benefit compared with no statin use (p < 0.005, hazard ratio [HR] = 0.60, 95% confidence interval [CI] = 0.39-0.94). This benefit remained after adjusting for other significant predictors of survival (p < 0.05, HR = 0.60, 95%CI = 0.39-0.94). A significant interaction effect was noted with bisoprolol, survival being greatest in the statin/bisoprolol group (p < 0.001, HR = 0.14, 95% CI = 0.03-0.60). Survival was 98.3% in the statin/bisoprolol group, 82.1% in the statin/placebo group, 87.2% in the no statin/bisoprolol group and 82.8% in the no statin/placebo group. The statin/bisoprolol group was also associated with fewer cardiovascular (p < 0.005) and sudden deaths (p < 0.0005) compared with other groups. CONCLUSIONS Despite the post-hoc, non-randomised nature of this analysis, these observations suggest that statin use appears to be beneficial in CHF. Furthermore, there appears to be a favourable interaction between statins and beta-blockade within the Second Cardiac Insufficiency Bisoprolol Study cohort. Prospective studies of statins are required to definitively address the role of these agents in established CHF.
Collapse
Affiliation(s)
- Henry Krum
- NHMRC Centre of Clinical Research Excellence in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia.
| | | | | | | | | | | | | |
Collapse
|
67
|
Eduardo Carreño J, Apablaza F, Paz Ocaranza M, E. Jalil J. Hipertrofia cardiaca: eventos moleculares y celulares. Rev Esp Cardiol 2006. [DOI: 10.1157/13087900] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
68
|
Carreño JE, Apablaza F, Ocaranza MP, Jalil JE. Cardiac Hypertrophy: Molecular and Cellular Events. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/s1885-5857(06)60796-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
69
|
Laufs U, Custodis F, Böhm M. HMG-CoA reductase inhibitors in chronic heart failure: potential mechanisms of benefit and risk. Drugs 2006; 66:145-54. [PMID: 16451090 DOI: 10.2165/00003495-200666020-00002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
HMG-CoA reductase inhibitors (statins) have been shown to reduce mortality and cardiovascular morbidity in patients with hyperlipidaemia and those with coronary artery disease. However, evidence for statin treatment in patients with chronic heart failure (CHF) remains a subject of debate. Patients with heart failure were generally excluded in the existing trials and a different patient population with a distinct pattern of morbidity and treatment was studied. In addition, no safety data are available for statins in patients with heart failure, where there are potential concerns about coenzyme Q10 depletion and excessive low-density lipoprotein reduction. This review summarises the clinical and preclinical evidence for potential beneficial effects of statins in CHF. In experimental systems, statins have been shown to improve cardiac function through antioxidative and anti-inflammatory action. Statins improve endothelial function, may reduce neurohormonal activation, and stimulate endothelial progenitor cells. Some of these effects occur independently of cholesterol lowering and can be explained by inhibition of isoprenylation of signal transducing proteins of the family of Rho guanosine triphosphatases. Two ongoing controlled randomised trials (CORONA [Controlled Rosuvastatin Multinational Study in Heart Failure] and GISSI-HF [Gruppo Italiano per lo Studio della Sopravvivenza nell'Infarto Miocardico--Heart Failure]) will help us to assess whether the described beneficial effects of statins in heart failure outweigh the potential negative effects and translate into the reduction of clinical endpoints.
Collapse
Affiliation(s)
- Ulrich Laufs
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany.
| | | | | |
Collapse
|
70
|
Cirrincione GM, Boluyt MO, Hwang HS, Bleske BE. 3-HMG-Coenzyme A Reductase Inhibition and Extracellular Matrix Gene Expression in the Pressure-Overloaded Rat Heart. J Cardiovasc Pharmacol 2006; 47:521-30. [PMID: 16680065 DOI: 10.1097/01.fjc.0000211745.70831.75] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The purpose of this study was to determine whether 3-HMG-Coenzyme A (HMG-CoA) reductase inhibition would attenuate the early pressure overload-induced activation of extracellular matrix genes in the left ventricle (LV) of the heart. Sprague-Dawley rats were randomized to 1 of 4 treatment groups: sham-operation+vehicle (SH-V), aortic constriction+vehicle (AC-V), AC+rosuvastatin (RSV, 2 mg/kg; AC-LO), and AC+RSV (10 mg/kg; AC-HI). Rats were injected with normal NaCl (V) or RSV once daily, beginning 1 day before surgery, and killed 1 or 3 days after surgery. Hemodynamic measurements were made in the open-chest anesthetized state. LV levels of transforming growth factor beta1 (TGF-beta1), procollagen 1 (C1), and fibronectin (FN) mRNA were measured by Northern blotting. AC induced a approximately 25% increase in LV weight after 3 days that was not altered by RSV treatment. LV expression of TGF-beta1, C1, and FN mRNA was approximately 2-fold, approximately 2.5-fold, and approximately 5-fold greater, respectively, in hearts of AC-V compared to SH-V rats 3 days post-operation, and was not significantly decreased by either dose of RSV. Inhibition of HMG-CoA reductase does not attenuate the pronounced aortic constriction-induced increases in the early expression of TGF-beta1, C1, and FN in this model of acute pressure overload of the rat heart.
Collapse
|
71
|
Hu TP, Xu FP, Li YJ, Luo JD. Simvastatin inhibits leptin-induced hypertrophy in cultured neonatal rat cardiomyocytes. Acta Pharmacol Sin 2006; 27:419-22. [PMID: 16539841 DOI: 10.1111/j.1745-7254.2006.00300.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
AIM To test the hypothesis that statins inhibit leptin-induced hypertrophy in cultured neonatal rat cardiomyocytes. METHODS Cultured neonatal rat cardiomyocytes were used to evaluate the effects of simvastatin on leptin-induced hypertrophy. Intracellular reactive oxygen species (ROS) levels were determined by using 2',7'-dichlorofluorescein diacetate (DCF-DA) fluorescence. Total intracellular RNA and cell protein content, which serve as cell proliferative markers, were assayed by using propidium iodide (PI) fluorescence and the Bio-Rad DC protein assay, respectively. The cell surface area, an indicator of cell hypertrophy, was quantified by using Leica image analysis software. RESULTS After 72 h treatment, leptin markedly increased RNA levels, cell surface area, and total cell protein levels in cardiomyocytes, which were significantly inhibited by simvastatin or catalase treatment. ROS levels were significantly elevated in cardiomyocytes treated with leptin for 4 h compared with those cells without leptin treatment. The increase in ROS levels in cardiomyocytes induced by leptin was reversed by treatment with simvastatin and catalase. CONCLUSION Simvastatin inhibits leptin-induced ROS-mediated hypertrophy in cultured neonatal rat cardiac myocytes. Statin therapy may provide an effective means of improving cardiac dysfunction in obese humans.
Collapse
Affiliation(s)
- Tai-Ping Hu
- Department of Pharmacology, Guangzhou Medical College, Guangzhou 510182, China
| | | | | | | |
Collapse
|
72
|
Abstract
Cardiac hypertrophy is a physiological adaptive response by the heart to pressure overload. However, after prolonged periods, this initial adaptive response becomes maladaptive, leading to increased mortality and morbidity from heart failure. Recently, 3-hydroxyl-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, or statins, have been shown to inhibit cardiac hypertrophy by cholesterol-independent mechanisms. Statins block the isoprenylation and activation of members of the Rho guanosine triphosphatase (GTPase) family, such as RhoA and Rac1. Since Rac1 is a requisite component of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, which is a major source of reactive oxygen species (ROS) in cardiovascular cells, the ability of statins to inhibit Rac1-mediated oxidative stress makes an important contribution to their inhibitory effects on cardiac hypertrophy.
Collapse
Affiliation(s)
- Hironori Nakagami
- Vascular Medicine Research, Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | | |
Collapse
|
73
|
Wu L, Zhao L, Zheng Q, Shang F, Wang X, Wang L, Lang B. Simvastatin attenuates hypertrophic responses induced by cardiotrophin-1 via JAK–STAT pathway in cultured cardiomyocytes. Mol Cell Biochem 2006; 284:65-71. [PMID: 16534557 DOI: 10.1007/s11010-005-9014-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Accepted: 09/21/2005] [Indexed: 11/28/2022]
Abstract
UNLABELLED Cardiotrophin-1 (CT-1) is a cytokine involved in the growth and survival of cardiac cells via activation of the Janus activated kinase/signal transducer activator of transcription (JAK/STAT). Statins, 3-Hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, have effects that extend beyond cholesterol reduction and inhibit vascular smooth muscle cell (VSMC) proliferation and cardiac hypertrophy. However, whether stains also can inhibitin vitromyocardial hypertrophy or not still remains elusive. The purpose of this study was to explore the effects of simvastatin on the hypertrophy of cultured rat cardiomyocytes induced by CT-1 and to investigate whether this effect was mediated via JAK-STAT signaling pathway. METHODS AND RESULTS Primary cardiomyocytes from 2-day-old (P2) rats were cultured, stimulated with CT-1, and treated with various concentration of simvastatin. Incorporation of [(3)H] leucine, reverse transcription-polymerase chain reaction and western blotting techniques were used to investigate cardiacmyocyte size, ANP mRNA and JAK-STAT protein expression. Simvastatin was proved, in a dose-independent manner, to decrease cardiacmyocytes size as well as protein synthesis, and inhibit ANP mRNA synthesis and JAK-STAT protein expression induced by CT-1 in cardiacmyocytes. CONCLUSION These results suggest that simvastatin can ameliorate cardiacmyocytes hypertrophyin vitrovia JAK-STAT signaling pathways. The present study provides a novel understanding and alternative therapeutic strategy for cardiac hypertrophy.
Collapse
Affiliation(s)
- LiJun Wu
- Institute Hypertension, Department of Cardiology, TangDu Hospital, Fourth Military Medical University, Xi'an 710038, PR China.
| | | | | | | | | | | | | |
Collapse
|
74
|
Shibata S, Nagase M, Fujita T. Fluvastatin Ameliorates Podocyte Injury in Proteinuric RatsviaModulation of Excessive Rho Signaling. J Am Soc Nephrol 2006; 17:754-64. [PMID: 16452496 DOI: 10.1681/asn.2005050571] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Statins have been reported to confer renoprotection in several experimental models of renal disease through pleiotropic actions. The roles of statins in glomerular podocytes have not been explored. The objective of this study was to evaluate the effects of fluvastatin on podocyte and tubulointerstitial injury in puromycin aminonucleoside (PAN)-induced nephrosis. PAN induced massive proteinuria and serum creatinine elevation on day 7, which were significantly suppressed by fluvastatin. Immunofluorescence studies of podocyte-associated proteins nephrin and podocin revealed diminished and discontinuous staining patterns in rats with PAN nephrosis, indicating severe podocyte injury. Fluvastatin treatment dramatically mitigated the abnormal staining profiles. Reduction of nephrin expression by PAN and its reversal by fluvastatin were confirmed by quantitative analyses. By electron microscopy, effacement of foot processes was ameliorated in fluvastatin-treated rats. Fluvastatin also mitigated tubulointerstitial damage in PAN nephrosis, with the repression of PAN-induced NF-kappaB and activator protein-1 activation in the kidneys. In addition, expression of activated membrane-bound small GTPase RhoA was markedly increased in the glomeruli of PAN nephrosis, which was inhibited by fluvastatin treatment. In cultured podocytes, fluvastatin suppressed PAN-evoked activation of RhoA and actin cytoskeletal reorganization. Furthermore, fasudil, a specific Rho-kinase inhibitor, successfully ameliorated PAN-induced podocyte damage and proteinuria. In summary, fluvastatin alleviated podocyte and tubulointerstitial injury in PAN nephrosis. The beneficial effects of fluvastatin on podocytes can be attributable to direct modulation of excessive RhoA activity. Our data suggest a therapeutic role for statins in clinical conditions that are relevant to podocyte injury.
Collapse
Affiliation(s)
- Shigeru Shibata
- Department of Nephrology and Endocrinology, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | | | | |
Collapse
|
75
|
Abstract
PURPOSE OF REVIEW The 3-hydroxy-3-methylglutaryl-coenzyme-A reductase inhibitors, or statins, have been shown to reduce cardiovascular morbidity and mortality among a wide spectrum of patients with established atherosclerotic vascular disease. Mounting experimental and clinical evidence also suggest a potential benefit as well as theoretical harm of statin therapy in patients with heart failure. RECENT FINDINGS This article briefly summarizes the therapeutic properties of statins that may be of benefit to patients with heart failure and the theoretical adverse effects of cholesterol reduction in this group of patients. A number of nonrandomized clinical studies over the past several years have shown an association between statin use and reduced overall mortality. Several large-scale randomized studies designed to confirm these findings are currently under way. SUMMARY Statin therapy appears to improve clinical outcomes in patients with both ischemic and nonischemic cardiomyopathy independently of their cholesterol-lowering properties. The theoretical adverse properties of statins in heart failure patients have not been substantiated in small to medium-sized clinical trials. Although the encouraging results of these preliminary studies suggest a role for statin therapy in heart failure, larger studies are needed to validate these findings. Several ongoing randomized trials are currently under way to evaluate the effect of statin therapy on cardiovascular outcomes in heart failure patients. The results of these studies, expected in the next several years, should provide scientific evidence for the role of statins in the treatment of failure.
Collapse
Affiliation(s)
- Michael Pham
- Division of Cardiovascular Medicine, Stanford University School of Medicine and Cardiology Section, VA Palo Alto Health Care System, Palo Alto, California, USA
| | | | | |
Collapse
|
76
|
Sorrentino S, Landmesser U. Nonlipid-lowering effects of statins. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2005; 7:459-66. [PMID: 16283973 DOI: 10.1007/s11936-005-0031-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Statins have been shown to effectively reduce cardiovascular events in patients with hypercholesterolemia, diabetes, and coronary disease, and after an acute coronary syndrome in several large-scale clinical trials. Interestingly, numerous studies have suggested that statins exert potentially important effects independent of lipid lowering (ie, improve endothelial function, reduce oxidant stress), and have direct anti-inflammatory, antithrombotic, and plaque-stabilizing effects. These beneficial effects may contribute to cardiovascular protection by statin therapy beyond low-density lipoprotein (LDL) cholesterol lowering. Therefore, it remains unclear at present to what extent the beneficial cardiovascular effects of statin treatment are dependent on LDL cholesterol lowering (ie, whether the same effect would be achieved by other modes of lipid lowering). Consequently, statins should be used as a first-line therapy for lipid lowering. Importantly, the observation of LDL cholesterol-independent effects of statins has stimulated clinical studies testing a wider use of statin treatment for diseases that are not thought to be related to increased LDL cholesterol levels, such as in patients with chronic heart failure (in particular dilated cardiomyopathy) and even in inflammatory diseases such as rheumatoid arthritis and multiple sclerosis.
Collapse
Affiliation(s)
- Sajoscha Sorrentino
- Medizinische Hochschule Hannover, Abteilung Kardiologie und Angiologie, Carl Neuberg Str.1, Hannover 30625, Germany
| | | |
Collapse
|
77
|
Takebayashi K, Matsumoto S, Wakabayashi S, Inukai Y, Matsutomo R, Aso Y, Inukai T. The effect of low-dose atorvastatin on circulating monocyte chemoattractant protein-1 in patients with type 2 diabetes complicated by hyperlipidemia. Metabolism 2005; 54:1225-9. [PMID: 16125534 DOI: 10.1016/j.metabol.2005.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2004] [Accepted: 04/11/2005] [Indexed: 10/25/2022]
Abstract
The effect of low-dose atorvastatin on various biomarkers was investigated in patients with type 2 diabetes complicated by hyperlipidemia. At 0 and 12 weeks in both the atorvastatin group (10 mg/d; n=17) and the no-drug group (n=10), high-sensitivity C-reactive protein (hsCRP), monocyte chemoattractant protein (MCP)-1, plasminogen activator inhibitor (PAI)-1, and fibrinogen were measured. At baseline, the entire group of diabetic patients (n=27) had significantly higher values of hsCRP and fibrinogen compared with those in age-matched healthy subjects (n=29): 0.801 (0.306, 1.760) vs 0.282 (0.143, 0.6505) mg/L, P=.0042; 329.1+/-55.0 vs 212.4+/-35.9 mg/dL, P<.0001, respectively. High-sensitivity C-reactive protein decreased significantly with atorvastatin treatment, from 0.801 (0.243, 1.865) to 0.308 (0.200, 0.804) mg/L (P=.0191). Although MCP-1, PAI-1, and fibrinogen did not decrease in the atorvastatin patients overall, the decrease of MCP-1 was significant in women (n=10; from 241.9+/-45.8 to 215.4+/-49.5 pg/mL, P=.0332). No correlation was found between changes in the serum lipid concentrations and changes in hsCRP, MCP-1, PAI-1, or fibrinogen in either the atorvastatin or the no-drug group. In conclusion, low-dose atorvastatin (10 mg/d) significantly decreased hsCRP in patients overall, and MCP-1 was also decreased in women. These findings suggest the possibility that atorvastatin provides an anti-inflammatory effect even at a low dose.
Collapse
Affiliation(s)
- Kohzo Takebayashi
- Department of Medicine, Koshigaya Hospital, Dokkyo University School of Medicine, Koshigaya 343-8555, Japan.
| | | | | | | | | | | | | |
Collapse
|
78
|
González A, López B, Díez J. New directions in the assessment and treatment of hypertensive heart disease. Curr Opin Nephrol Hypertens 2005; 14:428-34. [PMID: 16046900 DOI: 10.1097/01.mnh.0000174143.30045.bd] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW This article will review briefly the emerging new concepts in the diagnosis and treatment of myocardial fibrosis in patients with hypertensive heart disease. RECENT FINDINGS Although hypertensive heart disease is characterized clinically by development of left-ventricular hypertrophy in the absence of a cause other than arterial hypertension, changes in the composition of myocardial tissue also develop in arterial hypertension leading to structural remodeling of the myocardium (e.g. fibrosis). Recent available data on the determination of serum concentrations of collagen-derived serum peptides and quantitative analysis of echoreflectivity to address the presence of fibrosis in the myocardium of hypertensive patients are promising. In addition, preliminary data suggest that the goal of reducing myocardial fibrosis is achievable in patients with hypertensive heart disease using specific antihypertensive agents (e.g. those interfering with angiotensin II). SUMMARY The time has come to revisit the current management of hypertensive heart disease simply focused on detecting left-ventricular hypertrophy and controlling blood pressure to reduce left-ventricular mass. It is necessary to develop new approaches aimed at assessing and repairing alterations of myocardial structure and protect myocardial function and, in so doing, to reduce in a more-effective manner adverse risk associated with hypertensive heart disease.
Collapse
Affiliation(s)
- Arantxa González
- Division of Cardiovascular Pathophysiology, Centre for Applied Medical Research, University Clinic, School of Medicine, University of Navarra, C/Pio XII 55, 31008 Pamplona, Spain
| | | | | |
Collapse
|
79
|
Abstract
Cardiac hypertrophy and heart failure are leading causes of morbidity and mortality worldwide. The 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, or statins, have been shown to inhibit cardiac hypertrophy and improve symptoms of heart failure by cholesterol-independent mechanisms. Statins block the isoprenylation and function of members of the Rho GTPase family, such as Rac1 and RhoA. Because Rac1 is a requisite component of NADPH oxidase, which is a major source of reactive oxygen species in cardiovascular cells, the ability of statins to inhibit Rac1-mediated oxidative stress contributes importantly to their inhibitory effects on cardiac hypertrophy. Furthermore, inhibition of RhoA by statins leads to the activation of protein kinase B/Akt and upregulation of Type 3 nitric oxide synthase in the endothelium and the heart. This activation and upregulation results in increased angiogenesis and myocardial perfusion, decreased myocardial apoptosis, and improvement in endothelial and cardiac function. Because these effects of statins occur independent of cholesterol lowering, statins may have therapeutic benefits in nonhyperlipidemic patients with cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Seema Mital
- Division of Pediatric Cardiology, Columbia University, College of Physicians and Surgeons, New York, New York
| | | |
Collapse
|
80
|
Keller KB, Lemberg L. Statin Therapy in Congestive Heart Failure. Am J Crit Care 2005. [DOI: 10.4037/ajcc2005.14.4.338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Kathryn Buchanan Keller
- The Florida Atlantic University Christine E. Lynn College of Nursing, Boca Raton, Fla (kbk), and the Division of Cardiology, Department of Medicine, University of Miami School of Medicine, Miami, Fla (ll)
| | - Louis Lemberg
- The Florida Atlantic University Christine E. Lynn College of Nursing, Boca Raton, Fla (kbk), and the Division of Cardiology, Department of Medicine, University of Miami School of Medicine, Miami, Fla (ll)
| |
Collapse
|
81
|
Cassis LA, Helton MJ, Howatt DA, King VL, Daugherty A. Aldosterone does not mediate angiotensin II-induced atherosclerosis and abdominal aortic aneurysms. Br J Pharmacol 2005; 144:443-8. [PMID: 15655500 PMCID: PMC1576022 DOI: 10.1038/sj.bjp.0706098] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
We have demonstrated previously that infusion of angiotensin II (AngII) into hyperlipidemic mice augments atherosclerosis and results in the formation of abdominal aortic aneurysms (AAA). The purpose of this study was to determine the role of aldosterone in these AngII-induced vascular pathologies. Male apolipoprotein E-/- (apoE) mice were infused with either vehicle or aldosterone (50 or 200 ng kg(-1) min(-1)). Arterial blood pressure was determined throughout the study and serum lipid concentrations and vascular pathology were quantified after 28 days of infusion. Infusion of aldosterone did not influence body weight or serum cholesterol concentrations. Kidney weight was increased dose-dependently by aldosterone infusion. Systolic blood pressure was not significantly altered by aldosterone. Plasma aldosterone concentrations were increased dose-dependently by infusion of aldosterone. However, there was no effect of aldosterone on the extent of atherosclerosis and AAAs were not formed. Implantation of pellets containing spironolactone (16 mg kg(-1) day(-1)) in AngII-infused apoE-/- mice (1000 ng kg(-1) min(-1)) had no effect on AngII-induced elevations in blood pressure. Plasma aldosterone concentration was not influenced by coadministration of spironolactone with AngII. Spironolactone administration did not influence the extent of atherosclerosis. Moreover, spironolactone had no significant effect on AngII-induced AAA (incidence of AAA formation: 80 versus 70% for vehicle versus spironolactone, respectively; not significant). These studies demonstrate that the AngII-induced vascular pathologies of atherosclerosis and AAA formation are not mediated through aldosterone.
Collapse
Affiliation(s)
- Lisa A Cassis
- Graduate Center for Nutritional Sciences, Charles T. Wethington Building, University of Kentucky, Lexington, KY 40536, U.S.A.
| | | | | | | | | |
Collapse
|
82
|
Gerbaulet SP, Krämer J, Bohlender J, Dietz R, Gross CM. Blood Pressure-Independent ET A and AT 1 Receptor Blocker Effects on the Coronaries of Rats Harboring Human Renin and Angiotensinogen Genes. Kidney Blood Press Res 2005; 28:134-43. [PMID: 15915003 DOI: 10.1159/000085956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Blood pressure-independent (BP) effects of angiotensin (Ang) II and endothelin (ET) on coronaries (remodeling) in high renin hypertension are incompletely understood. METHODS We studied the effects of subdepressor doses of Ang II receptor (AT1) blockade with losartan (10 mg/kg/day gavage) and endothelin A receptor (ETA) blockade with LU135252 (30 mg/kg/day) on the coronaries of rats harboring human renin and angiotensinogen genes (dTGR). Nontransgenic Sprague-Dawley rats were controls. The rats were treated between the ages of 6 and 10 weeks. Coronary cross-sectional area [CSA; 0.79 x (external diameter2 - internal diameter2)], cell proliferation, and infiltration of monocytes/macrophages were determined. RESULTS Monotherapy did not lower BP while combination treatment did (p < 0.05). All treatments reduced mortality (p < 0.01). CSA was decreased by all treatments compared to vehicle, independent of blood pressure (p < 0.05). Extensive proliferation by PCNA staining and infiltration of ED-1-positive cells was diminished by both treatment and the combination. CONCLUSIONS The data show that Ang II promotes coronary inflammation and remodeling, in part independent of blood pressure but dependent upon ET signaling. Combination treatment directed at both pathways may improve outcome, independent of blood pressure reduction.
Collapse
Affiliation(s)
- Stephan P Gerbaulet
- Franz Volhard Clinic, HELIOS Klinikum-Berlin and Max Delbrück Center, Medical Faculty of the Charité, Humboldt University of Berlin, Germany.
| | | | | | | | | |
Collapse
|
83
|
Abstract
Statins are potent inhibitors of cholesterol biosynthesis. In clinical trials, statins are beneficial in the primary and secondary prevention of coronary heart disease. However, the overall benefits observed with statins appear to be greater than what might be expected from changes in lipid levels alone, suggesting effects beyond cholesterol lowering. Indeed, recent studies indicate that some of the cholesterol-independent or "pleiotropic" effects of statins involve improving endothelial function, enhancing the stability of atherosclerotic plaques, decreasing oxidative stress and inflammation, and inhibiting the thrombogenic response. Furthermore, statins have beneficial extrahepatic effects on the immune system, CNS, and bone. Many of these pleiotropic effects are mediated by inhibition of isoprenoids, which serve as lipid attachments for intracellular signaling molecules. In particular, inhibition of small GTP-binding proteins, Rho, Ras, and Rac, whose proper membrane localization and function are dependent on isoprenylation, may play an important role in mediating the pleiotropic effects of statins.
Collapse
Affiliation(s)
- James K. Liao
- Vascular Medicine Research, Brigham & Women’s Hospital, Cambridge, Massachusetts 02139;
| | - Ulrich Laufs
- Klinik Innere Medizin III, Universität des Saarlandes, 66421 Homburg, Germany;
| |
Collapse
|
84
|
Böhm M, Hjalmarson A, Kjekshus J, Laufs U, McMurray J, van Veldhuisen DJ. Heart failure and statins—Why do we need a clinical trial? ACTA ACUST UNITED AC 2005; 94:223-30. [PMID: 15803258 DOI: 10.1007/s00392-005-0210-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2004] [Accepted: 11/16/2004] [Indexed: 11/30/2022]
Abstract
The effect of statins to reduce mortality and morbidity in primary and secondary prevention as well as in acute coronary syndrome is well established. Recent data show that pleiotropic effects might also have direct effects on the myocardial cell. However, in chronic heart failure the outcome is inversely related to LDL-plasma concentrations and other pleiotropic effects might impair mitochondrial function. Since there are no safety data on the use of statins in chronic heart failure, a controlled randomized and placebo-controlled trial is urgently needed.
Collapse
Affiliation(s)
- M Böhm
- Klinik für Innere Medizin III, Universitätsklinik des Saarlandes, Kirrberger Strasse, 66421 Homburg, Gemany.
| | | | | | | | | | | |
Collapse
|
85
|
Goikoetxea MJ, Beaumont J, Díez J. Peroxisome proliferator-activated receptor alpha and hypertensive heart disease. Drugs 2005; 64 Suppl 2:9-18. [PMID: 15765888 DOI: 10.2165/00003495-200464002-00003] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Peroxisome proliferator-activated receptor alpha (PPARalpha) is a ligand-activated transcription factor belonging to the nuclear hormone receptor superfamily. It is expressed by cardiomyocytes and regulates gene expression of key proteins involved in myocardial lipid and energy metabolism. Accordingly, the activitity of PPARalpha is an important determinant of cardiomyocyte lipid homeostasis and ATP production. Currently, animal and human data suggest that deactivation of PPARalpha may contribute substantially to phenotypic changes that accompany cardiac growth in conditions of pressure overload, and the hypothesis emerges that a compromised PPARalpha activity may participate in the transition from compensated left ventricular hypertrophy to heart failure in hypertensive heart disease. The availability of PPARalpha activators (e.g. fibric acid derivates and statins) must stimulate investigation into the potential cardioprotective actions of these compounds beyond their hypolipidaemic effects and via restoration of PPARalpha activity in the hypertrophied and failing heart.
Collapse
Affiliation(s)
- Maria J Goikoetxea
- Area of Cardiovascular Pathophysiology, Centre for Applied Medical Research, University Clinic, School of Medicine, University of Navarra, Pamplona, Spain
| | | | | |
Collapse
|
86
|
Koh KK, Son JW, Ahn JY, Jin DK, Kim HS, Kim DS, Han SH, Chung WJ, Park GS, Shin EK. Vascular effects of simvastatin combined with ramipril in hypercholesterolemic patients with coronary artery disease, compared with simvastatin alone: a randomized, double-blind, placebo-controlled, crossover study. Atherosclerosis 2005; 177:147-53. [PMID: 15488877 DOI: 10.1016/j.atherosclerosis.2004.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2004] [Revised: 06/14/2004] [Accepted: 06/30/2004] [Indexed: 11/24/2022]
Abstract
Because the mechanisms of the biological effects of statin and angiotensin converting enzyme inhibitor therapies differ, we studied the vascular responses to these therapies in hypercholesterolemic patients with coronary artery disease. We administered simvastatin 20 mg and placebo or ramipril 10 mg daily during 2 months with washout 2 months to 32 hypercholesterolemic patients with coronary artery disease. This study was randomized, double-blind, placebo-controlled, crossover in design. Simvastatin alone or combined with ramipril significantly changed lipoproteins, and improved the percent flow-mediated dilator response to hyperemia relative to baseline measurements by 33 +/- 6% and by 50 +/- 14%, respectively (both P <0.001) and reduced plasma levels of nitrate relative to baseline measurements (P=0.413 and 0.037, respectively), the plasma MDA levels relative to baseline measurements by 8 +/- 8% and by 18 +/- 9% (P=0.039 and P <0.001, respectively) and MCP-1 relative to baseline measurements by 7 +/- 4% and by 13 +/- 3%, respectively (P=0.019 and P <0.001, respectively), and CRP from 0.22 to 0.14 mg/dl and from 0.22 to 0.15 mg/dl, respectively (P=0.124 and 0.002, respectively), and PAI-1 antigen relative to baseline measurements (P=0.690 and 0.018, respectively). However, simvastatin combined with ramipril changed to greater but statistically insignificant extent the percent flow-mediated dilator response to hyperemia and plasma levels of nitrate, MDA, MCP-1, and PAI-1 antigen than simvastatin alone. Simvastatin alone or combined with ramipril showed significant beneficial effects on endothelial function in hypercholesterolemic patients with coronary artery disease. However, simvastatin combined with ramipril did not significantly change, compared with simvastatin alone.
Collapse
Affiliation(s)
- Kwang Kon Koh
- kVascular Medicine and Atherosclerosis Unit, Cardiology, Gil Heart Center, Gachon Medical School, Kuwol-dong 1198, Namdong-gu, Incheon 405-760, Korea.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Chen Y, Ross BM, Currie RW. Heat shock treatment protects against angiotensin II-induced hypertension and inflammation in aorta. Cell Stress Chaperones 2005; 9:99-107. [PMID: 15270082 PMCID: PMC1065311 DOI: 10.1379/csc-1r.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Angiotensin II (Ang II) is a potent vasoconstrictor and induces inflammation and end-organ injury through its activation of the proinflammatory transcription factor, nuclear factor-kappaB (NF-kappaB). Heat shock (HS) treatment with subsequent expression of heat shock proteins (Hsps) is an effective strategy for tissue protection against oxidative injuries. Recently, HS and Hsps have been shown to interact with NF-kappaB in tissue injury. In this study, we investigated whether HS could protect against Ang II-induced hypertension and inflammation by inhibiting NF-kappaB. Sprague-Dawley rats were divided into control and HS groups. Control and 24-hour post-heat shocked rats were treated with Ang II. At days 1, 3, 5, 7, 11, and 14 after Ang II administration, systolic blood pressures were measured by tail-cuff plethysmography, and aorta tissues were collected. Aorta NF-kappaB deoxyribonucleic acid-binding activity was measured by electrophoretic mobility shift assay, and NF-kappaB p65 subunit, Hsp70, Hsp27, and interleukin-6 (IL-6) expressions were measured by Western analysis. HS treatment significantly decreased Ang II-induced hypertension. The activation of NF-kappaB in aorta by Ang II was suppressed by HS treatment. The elevated expression of IL-6 induced by Ang II treatment was also decreased by HS treatment. Although Ang II treatment induced an increase in Hsp70 and Hsp27, HS treatment induced a greater elevation of Hsp70 and Hsp27 expression. HS treatment protects against Ang II-induced hypertension and inflammation. This protection may relate to the interaction of Hsps and the NF-kappaB pathway.
Collapse
Affiliation(s)
- Yu Chen
- Department of Anatomy & Neurobiology, Dalhousie University, 5850 College Street, Halifax, Nova Scotia, Canada B3H 1X5
| | | | | |
Collapse
|
88
|
Abstract
Cardiac hypertrophy leading to heart failure is a major cause of morbidity and mortality worldwide. The 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, or statins, have been shown to inhibit cardiac hypertrophy and improve symptoms of heart failure by cholesterol-independent mechanisms. Statins block the isoprenylation and function of members of the Rho guanosine triphosphatase family, such as Rac1 and RhoA. Because Rac1 is a requisite component of reduced nicotinamide adenine dinucleotide phosphate oxidase, which is a major source of reactive oxygen species in cardiovascular cells, the ability of statins to inhibit Rac1-mediated oxidative stress contributes importantly to their inhibitory effects on cardiac hypertrophy. Furthermore, inhibition of RhoA by statins leads to the activation of protein kinase B/Akt and up-regulation of endothelial nitric oxide synthase in the endothelium and the heart. This results in increased angiogenesis and myocardial perfusion, decreased myocardial apoptosis, and improvement in endothelial and cardiac function. Because these effects of statins occur independently of cholesterol lowering, statins may have therapeutic benefits in nonhyperlipidemic patients with cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- James K Liao
- Vascular Medicine Research, Department of Medicine, Brigham & Women's Hospital, Cambridge, MA 02139, USA.
| |
Collapse
|
89
|
Chen Y, Arrigo AP, Currie RW. Heat shock treatment suppresses angiotensin II-induced activation of NF-κB pathway and heart inflammation: a role for IKK depletion by heat shock? Am J Physiol Heart Circ Physiol 2004; 287:H1104-14. [PMID: 15087290 DOI: 10.1152/ajpheart.00102.2004] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heat shock (HS) proteins (Hsps) function in tissue protection through their chaperone activity and by interacting with cell signaling pathways to suppress apoptosis. Here, we investigated the effect of HS treatment on the nuclear factor (NF)-κB signaling pathway in the angiotensin II (ANG II) model of inflammation. Male Sprague-Dawley rats were divided into sham and HS-, ANG II-, and HS + ANG II-treated groups. HS treatment was administered 24 h before the initiation of ANG II infusion. HS treatment (42°C for 15 min) decreased 7-day ANG II-induced hypertension from 191 ± 4 to 147 ± 3 mmHg ( P < 0.01). Histological staining of hearts showed that HS treatment reduced ANG II-induced leukocyte infiltration, perivascular and interstitial inflammation, and fibrosis. Heart NF-κB nuclear translocation and activity, examined by Western blot analysis and electrophoretic mobility shift assay, was suppressed by HS treatment. HS treatment depleted IκB kinase-α (IKK-α) and phosphorylated IKK-α and suppressed the depletion of IκB-α and the accumulation of phosphorylated IκB-α. HS treatment blocked ANG II induced expression of IL-6 and ICAM-1 in the heart. ANG II and HS treatment induced high-level expression of Hsp27 and Hsp70 and their phosphorylation. Phosphorylated isoforms of Hsp27 and Hsp70 may play an important role in protecting the heart against ANG II-induced inflammation.
Collapse
Affiliation(s)
- Yu Chen
- Department of Anatomy and Neurobiology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 1X5
| | | | | |
Collapse
|
90
|
Tavazzi L, Tognoni G, Franzosi MG, Latini R, Maggioni AP, Marchioli R, Nicolosi GL, Porcu M. Rationale and design of the GISSI heart failure trial: a large trial to assess the effects of n-3 polyunsaturated fatty acids and rosuvastatin in symptomatic congestive heart failure. Eur J Heart Fail 2004; 6:635-41. [PMID: 15302013 DOI: 10.1016/j.ejheart.2004.03.001] [Citation(s) in RCA: 194] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2003] [Revised: 09/19/2003] [Accepted: 03/01/2004] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The GISSI Heart Failure project is a large-scale, randomized, double-blind study designed to investigate the effects of n-3 polyunsaturated fatty acids and rosuvastatin on mortality and morbidity in patients with symptomatic heart failure. METHODS AND RESULTS Patients with New York Heart Association classes II to IV heart failure, already receiving optimized recommended therapy, will be recruited in a nation-wide network of more than 300 cardiology and internal medicine services to be randomly allocated to treatment with n-3 polyunsaturated fatty acids (1 g daily) or the corresponding placebo. Patients with no clear indication or contraindication to cholesterol-lowering therapy will be further randomized to receive low-dose rosuvastatin (10 mg daily) or placebo. According to data available in heart failure registries, it is expected that 70% of the patients will be suitable to enter both components of the trial, which assume the same co-primary endpoints: (a) 15% reduction of all-cause mortality and (b) 20% reduction of all-cause mortality or cardiovascular hospitalizations. The trial is event-driven and will continue either until at least 1252 deaths have been recorded or a reduction of all-cause mortality will satisfy the significance boundaries, which have been established to stop the study. The recruitment of the planned sample size of approximately 7000 patients randomized in the n-3 PUFA trial is expected to be completed within 18 months from the trial start. As of February 29, 2004, 4624 heart failure patients have been included in the trial. CONCLUSION The GISSI-HF project, with its protocol articulated into two independent randomization schemes, has the aim and the power to verify the hypothesis that n-3 polyunsaturated fatty acids and rosuvastatin can favorably modify the prognosis of patients with symptomatic heart failure.
Collapse
|
91
|
Abstract
Cardiac hypertrophy leading to heart failure is a major cause of morbidity and mortality worldwide. The 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, or statins, have been shown to inhibit cardiac hypertrophy and improve symptoms of heart failure by cholesterol-independent mechanisms. Statins block the isoprenylation and function of members of the Rho guanosine triphosphatase family, such as Rad and RhoA. Because Rad is a requisite component of reduced nicotinamide adenine dinucleotide phosphate oxidase, which is a major source of reactive oxygen species in cardiovascular cells, the ability of statins to inhibit Rad-mediated oxidative stress contributes importantly to their inhibitory effects on cardiac hypertrophy. Furthermore, inhibition of RhoA by statins leads to the activation of protein kinase B/Akt and up-regulation of endothelial nitric oxide synthase in the endothelium and the heart. This results in increased angiogenesis and myocardial perfusion, decreased myocardial apoptosis, and improvement in endothelial and cardiac function. Because these effects of statins occur independently of cholesterol lowering, statins may have therapeutic benefits in nonhyperlipidemic patients with cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- James K. Liao
- Vascular Medicine Research, Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
92
|
Horwich TB, MacLellan WR, Fonarow GC. Statin therapy is associated with improved survival in ischemic and non-ischemic heart failure. J Am Coll Cardiol 2004; 43:642-8. [PMID: 14975476 DOI: 10.1016/j.jacc.2003.07.049] [Citation(s) in RCA: 304] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2003] [Revised: 07/02/2003] [Accepted: 07/28/2003] [Indexed: 11/25/2022]
Abstract
OBJECTIVES This study aimed to investigate the impact of hydroxymethylglutaryl coenzyme A reductase inhibitor (statin) therapy in patients with advanced heart failure (HF). BACKGROUND Although statins are known to reduce mortality in coronary artery disease (CAD), the impact of statin therapy in patients with HF has not been well studied. Both the potential risks and benefits of statins in HF have been described. METHODS We studied a cohort of 551 patients with systolic HF (left ventricular ejection fraction [EF] <or=40%) referred to a single university center for clinical management and/or transplant evaluation. Survival without the necessity of urgent heart transplantation was determined. RESULTS The patients' mean age was 52 +/- 13 years; mean EF was 25 +/- 7%. Forty-five percent of the cohort had CAD, and 45% were receiving statin therapy, including 73% and 22% of CAD and non-CAD patients with HF, respectively. Patients receiving statins were significantly older and more likely to be male, with higher rates of hypertension, diabetes, and smoking. The EF and cholesterol levels were similar between treated and non-treated patients. Statin use was associated with improved survival without the necessity of urgent transplantation in both non-ischemic and ischemic HF patients (91% vs. 72%, p < 0.001 and 81% vs. 63%, p < 0.001 at one-year follow-up, respectively). After risk adjustment for age, gender, CAD, cholesterol, diabetes, medications, hemoglobin, creatinine, and New York Heart Association functional class, statin therapy remained an independent predictor of improved survival (hazard ratio 0.41 95% confidence interval 0.18 to 0.94). CONCLUSIONS Statin therapy is associated with improved survival in patients with ischemic and non-ischemic HF. Randomized trials are needed for confirmation of a therapeutic benefit.
Collapse
Affiliation(s)
- Tamara B Horwich
- Ahmanson-UCLA Cardiomyopathy Center, Los Angeles, California 90095-1679, USA
| | | | | |
Collapse
|
93
|
Simko F, Matuskova J, Luptak I, Krajcirovicova K, Kucharska J, Gvozdjakova A, Babal P, Pechanova O. Effect of simvastatin on remodeling of the left ventricle and aorta in L-NAME-induced hypertension. Life Sci 2004; 74:1211-24. [PMID: 14697405 DOI: 10.1016/j.lfs.2003.07.032] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
3-Hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors have been shown to prevent or reverse hypertrophy of the LV in several models of left ventricular hypertrophy. The aim of the present study was to determine whether treatment with simvastatin can prevent hypertension, reduction of tissue nitric oxide synthase activity and left ventricular (LV) remodeling in NG-nitro-L-arginine methyl ester(L-NAME)-induced hypertension. Four groups of rats were investigated: control, simvastatin (10 mg/kg), L-NAME (40 mg/kg) and L-NAME + simvastatin (in corresponding doses). Animals were sacrificed and studied after 6 weeks of treatment. The decrease of NO-synthase activity in the LV, kidney and brain was associated with hypertension, LV hypertrophy and fibrosis development and remodeling of the aorta in the L-NAME group. Simvastatin attenuated the inhibition of NO-synthase activity in kidney and brain, partly prevented hypertension development and reduced the concentration of coenzyme Q in the LV. Nevertheless, myocardial hypertrophy, fibrosis and enhancement of DNA concentration in the LV, and remodeling of the aorta were not prevented by simultaneous simvastatin treatment in the L-NAME treated animals. We conclude that the HMG-CoA reductase inhibitor simvastatin improved nitric oxide production and partially prevented hypertension development, without preventing remodeling of the left ventricle and aorta in NO-deficient hypertension.
Collapse
Affiliation(s)
- Fedor Simko
- Department of Pathophysiology, School of Medicine, Comenius University, Sasinkova 4, 813 72, Bratislava, Slovak Republic.
| | | | | | | | | | | | | | | |
Collapse
|
94
|
Chen Y, Ross BM, William Currie R. Heat shock treatment protects against angiotensin II–induced hypertension and inflammation in aorta. Cell Stress Chaperones 2004. [DOI: 10.1379/1466-1268(2004)009<0099:hstpaa>2.0.co;2] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
95
|
Susic D, Varagic J, Ahn J, Slama M, Frohlich ED. Beneficial pleiotropic vascular effects of rosuvastatin in two hypertensive models. J Am Coll Cardiol 2003; 42:1091-7. [PMID: 13678936 DOI: 10.1016/s0735-1097(03)00926-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVES The goal of this research was to study the effects of rosuvastatin on systemic and regional hemodynamics in two hypertensive rat models, one genetic, the other induced with inhibition of nitric oxide synthesis. BACKGROUND Rats naturally have low cholesterol levels that are generally unaffected by statin therapy, thus providing a good model for studying cardiovascular effects unrelated to lipid metabolism. METHODS Male 20-week-old spontaneously hypertensive rats (SHR) were divided into five groups and given either vehicle or 1, 5, 10, and 20 mg/kg of rosuvastatin daily, by gavage, for 12 weeks. Wistar-Kyoto rats (WKY) were divided into four groups; the first received vehicle and the second rosuvastatin (20 mg/kg). The third and fourth groups were given N(omega)-nitro-L-arginine (L-NAME) (15 mg/kg/day) in drinking water, and the fourth group received rosuvastatin daily, 20 mg/kg for six weeks. At the end of the respective treatments, systemic and organ hemodynamics (radionuclide-labeled microspheres) and cardiovascular mass were determined in all rats. RESULTS Rosuvastatin reduced arterial pressure in SHR rats, but not in WKY/L-NAME rats. Total peripheral resistance decreased with rosuvastatin in both hypertensive models, whereas cardiac output increased with rosuvastatin in WKY/L-NAME rats. Neither cardiac nor aortic mass was changed. Regional hemodynamics improved with rosuvastatin in both hypertensive models, as evidenced by increased blood flows and decreased vascular resistances. No effect on plasma lipids was observed. CONCLUSIONS These results showed that rosuvastatin reduced arterial pressure in genetic hypertension and improved systemic and regional hemodynamics in both hypertensive models independently of cholesterol levels. Thus rosuvastatin improved systemic and regional hemodynamics by reducing vascular resistance.
Collapse
Affiliation(s)
- Dinko Susic
- Hypertension Research Laboratory, Ochsner Clinic Foundation, New Orleans, Louisiana 70121, USA.
| | | | | | | | | |
Collapse
|
96
|
Abstract
Cardiac hypertrophy is the heart's response to a variety of extrinsic and intrinsic stimuli that impose increased biomechanical stress. While hypertrophy can eventually normalize wall tension, it is associated with an unfavorable outcome and threatens affected patients with sudden death or progression to overt heart failure. Accumulating evidence from studies in human patients and animal models suggests that in most instances hypertrophy is not a compensatory response to the change in mechanical load, but rather is a maladaptive process. Accordingly, modulation of myocardial growth without adversely affecting contractile function is increasingly recognized as a potentially auspicious approach in the prevention and treatment of heart failure. In this review, we summarize recent insights into hypertrophic signaling and consider several novel antihypertrophic strategies.
Collapse
Affiliation(s)
- N Frey
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9148, USA.
| | | |
Collapse
|
97
|
Lazar HL, Bao Y, Zhang Y, Bernard SA. Pretreatment with statins enhances myocardial protection during coronary revascularization. J Thorac Cardiovasc Surg 2003; 125:1037-42. [PMID: 12771876 DOI: 10.1067/mtc.2003.177] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This experimental study was undertaken to determine whether pretreatment with statins would enhance myocardial protection and minimize ischemic injury during revascularization of acutely ischemic myocardium. METHODS In 20 pigs the second and third diagonal arteries were occluded for 90 minutes, followed by 45 minutes of blood cardioplegic arrest and 180 minutes of reperfusion. Ten pigs received atorvastatin (40 mg orally every day) for 21 days before surgical intervention; 10 others received no statins. Ischemic damage was assessed on the basis of the need for cardioversions for ventricular arrhythmias, regional wall-motion scores (4 = normal to -1 = dyskinesia) were determined by means of 2-dimensional echocardiography, endothelial function was assessed on the basis of bradykinin-induced coronary artery relaxation, and infarct size was calculated by determining the area of necrosis to the area of risk by means of histochemical staining. Results are given as means +/- SE. RESULTS Statin-treated animals required fewer cardioversions (0.11 +/- 0.01 vs 2.87 +/- 0.20, P =.0001), had improved wall-motion scores (2.81 +/- 0.10 vs 1.52 +/- 0.08, P =.01), had lower infarct size (21% +/- 2% vs 41% +/- 2%, P =.0001), and had more complete coronary artery relaxation (34% +/- 5% vs 8% +/- 4%, P =.01). Total serum cholesterol levels were similar between the groups (62 +/- 5 mg/dL for statin-treated animals vs 68 +/- 5 mg/dL for non-statin-treated animals, P =.30). CONCLUSIONS Pretreatment with statins enhances myocardial protection during revascularization by means of mechanisms that are independent of their cholesterol-lowering properties.
Collapse
Affiliation(s)
- Harold L Lazar
- Department of Cardiothoracic Surgery, The Boston Medical Center and The Boston University School of Medicine, Boston, MA 02118, USA.
| | | | | | | |
Collapse
|
98
|
Derosa G, Mugellini A, Ciccarelli L, Fogari R. Randomized, double-blind, placebo-controlled comparison of the action of orlistat, fluvastatin, or both an anthropometric measurements, blood pressure, and lipid profile in obese patients with hypercholesterolemia prescribed a standardized diet. Clin Ther 2003; 25:1107-22. [PMID: 12809960 DOI: 10.1016/s0149-2918(03)80070-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE The aim of this study was to assess obese patients with hypercholesterolemia whom were prescribed a standardized diet, comparing the action of orlistat, fluvastatin, orlistat with fluvastatin, and placebo on anthropometric measurements, blood pressure (BP), and lipid profile. METHODS This was a 1-year, randomized, double-blind, placebo-controlled trial. The patients were prescribed a controlled-energy diet and were randomly allocated to receive placebo, orlistat 120 mg TID (O group), fluvastatin 80 mg/d (F group), or olistat 120 mg TID with fluvastatin 80 mg/d (OF group). Clinical measurements (body weight, body mass index [BMI], waist circumference, and BP) and lipid profile assessment (total cholesterol [TC], low-density lipoprotein cholesterol [LDL-C], high-density lipoprotein cholesterol [HDL-C], and triglycerides [TGs]) were performed at baseline and after 6 months and 1 year of treatment. RESULTS The study included 99 obese patients with hypercholesterolemia (48 men and 51 women; mean [SD] age, 51 [9] years). There were no significant differences between groups in baseline demographic, BP, or plasma lipid values. Three patients dropped out (2 women in the O group and 1 man in the OF group) due to adverse events related to orlistat treatment, including gastrointestinal events (oily spotting and fecal urgency). Ninety-six patients completed the study. There were significant differences from baseline (mean [SD]) in BMI, waist circumference reduction (WCR), and body weight loss (BWL) at 6 months in the OF group (29.9 [1.1] kg/m(2), 2.7 [0.8] cm, and 7.4 [0.9] kg, respectively; all P < 0.05), and BMI, WCR, and BWL at 1 year in the O group (29.0 [1.0] kg/m(2), 3.0 [1.0] cm, and 8.6 [1.0] kg, respectively; all P < 0.02), the F group (29.3 [1.6] kg/m(2), 2.4 [1.0] cm, and 8/0 [1.0] kg, respectively; all P < 0.05), and the OF group (28.4 [0.6] kg/m(2), 4.0 [0.6] cm, and 11.4 [1.0] kg, respectively; all P < 0.01). Significant reductions from baseline in systolic and diastolic BP were observed at 1 year in the O and F groups (all P < 0.05) and the OF group (both P < 0.01). At 6 months, there were significant reductions from baseline in TC and LDL-C in the F group (both P < 0.05) and in TC, LDL-C, and TGs in the OF group (P < 0.02, P < 0.02, and P < 0.05, respectively), as well as a significant increase in HDL-C in the OF group (P < 0.02). At 1 year, there were significant reduction from baseline in TC in the O, F, and OF groups (P < 0.05 and P < 0.01, respectively), LDL-C (P < 0.05, P < 0.02, and P < 0.01, respectively), and TGs (P < 0.02, P < 0.05, and P < 0.02, respectively). Also at 1 year, HDL-C was significantly higher than baseline in the F and OF groups (P < 0.02 and P < 0.01, respectively). CONCLUSION Improvements in clinical and lipid-profile parameters were found at 1 year with all 3 treatments.
Collapse
Affiliation(s)
- Giuseppe Derosa
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy.
| | | | | | | |
Collapse
|
99
|
Nakagami H, Jensen KS, Liao JK. A novel pleiotropic effect of statins: prevention of cardiac hypertrophy by cholesterol-independent mechanisms. Ann Med 2003; 35:398-403. [PMID: 14572163 PMCID: PMC2662041 DOI: 10.1080/07853890310001294] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Cardiac hypertrophy is an initial physiological adaptive response by the heart to pressure overload. However, if pressure overload persists, frequently, the heart decompensates and develops 'pathophysiological' hypertrophy. This leads to increased mortality and morbidity and is an independent risk factor for heart failure. Because cardiac myocytes convert this pressure overload into intracellular biochemical signals, blocking this critical signaling pathway may be an important therapeutic target to prevent cardiac hypertrophy. Small GTP-binding proteins, in particular Rac1, have been suggested to play a key role in the development of cardiac hypertrophy. Recently, 3-hydroxyl-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, also called statins, have been shown to inhibit cardiac hypertrophy independent of their cholesterol lowering property. Statins block the isoprenylation and activation of members of the Rho family, such as RhoA and Rac1. Rac1 also regulates NADPH oxidase, which is a major source of reactive oxygen species (ROS) in cardiovascular cells. Growing evidence suggests that ROS may be involved in the process of cardiac hypertrophy and recent research has shown that statins attenuate oxidative stress through inhibition of Rac1. Overall, these pleiotropic effects of statins will give new insights into the process of cardiac hypertrophy.
Collapse
Affiliation(s)
- Hironori Nakagami
- Vascular Medicine Research Unit, Brigham & Women's Hospital and Harvard Medical School, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
100
|
Abstract
The introduction of the hydroxy methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (statins) in 1987 was a major advance in the prevention and treatment of cardiovascular disease. Several landmark clinical trials have demonstrated the benefit of lipid lowering with statins for the primary and secondary prevention of coronary heart disease (CHD), namely The Scandinavian Simvastatin Survival Study (4S), Cholesterol And Recurrent Events (CARE), Long-term Intervention with Pravastatin in Ischemic Disease (LIPID), West of Scotland Coronary Prevention Study (WOSCOPS) and Air Force/Texas Coronary Atherosclerosis Prevention Study (AFCAPS/TexCAPS). Although it is widely accepted that the majority of clinical benefit obtained with statins is a direct result of their lipid-lowering properties, these agents appear to display additional cholesterol-independent or pleiotropic effects on various aspects of cardiovascular disease, including improving endothelial function, decreasing vascular inflammation and enhancing plaque stability. Although the full impact of statin therapy on each of these processes is not fully understood, ongoing studies with current and new statins are likely to shed further light on the potential cholesterol-independent benefits of these agents.
Collapse
Affiliation(s)
- James K Liao
- Vascular Medicine Research, Brigham and Women's Hospital, 65 Landsdowne Street, Room 275, Cambridge, MA 02139, USA.
| |
Collapse
|