1
|
Thong EHE, Quek EJW, Loo JH, Yun CY, Teo YN, Teo YH, Leow AST, Li TYW, Sharma VK, Tan BYQ, Yeo LLL, Chong YF, Chan MY, Sia CH. Acute Myocardial Infarction and Risk of Cognitive Impairment and Dementia: A Review. BIOLOGY 2023; 12:1154. [PMID: 37627038 PMCID: PMC10452707 DOI: 10.3390/biology12081154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/05/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
Cognitive impairment (CI) shares common cardiovascular risk factors with acute myocardial infarction (AMI), and is increasingly prevalent in our ageing population. Whilst AMI is associated with increased rates of CI, CI remains underreported and infrequently identified in patients with AMI. In this review, we discuss the evidence surrounding AMI and its links to dementia and CI, including pathophysiology, risk factors, management and interventions. Vascular dysregulation plays a major role in CI, with atherosclerosis, platelet activation, microinfarcts and perivascular inflammation resulting in neurovascular unit dysfunction, disordered homeostasis and a dysfunctional neurohormonal response. This subsequently affects perfusion pressure, resulting in enlarged periventricular spaces and hippocampal sclerosis. The increased platelet activation seen in coronary artery disease (CAD) can also result in inflammation and amyloid-β protein deposition which is associated with Alzheimer's Dementia. Post-AMI, reduced blood pressure and reduced left ventricular ejection fraction can cause chronic cerebral hypoperfusion, cerebral infarction and failure of normal circulatory autoregulatory mechanisms. Patients who undergo coronary revascularization (percutaneous coronary intervention or bypass surgery) are at increased risk for post-procedure cognitive impairment, though whether this is related to the intervention itself or underlying cardiovascular risk factors is debated. Mortality rates are higher in dementia patients with AMI, and post-AMI CI is more prevalent in the elderly and in patients with post-AMI heart failure. Medical management (antiplatelet, statin, renin-angiotensin system inhibitors, cardiac rehabilitation) can reduce the risk of post-AMI CI; however, beta-blockers may be associated with functional decline in patients with existing CI. The early identification of those with dementia or CI who present with AMI is important, as subsequent tailoring of management strategies can potentially improve outcomes as well as guide prognosis.
Collapse
Affiliation(s)
- Elizabeth Hui En Thong
- Internal Medicine Residency, National University Health System, Singapore 119074, Singapore; (E.H.E.T.); (Y.H.T.); (A.S.T.L.)
| | - Ethan J. W. Quek
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
| | - Jing Hong Loo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
| | - Choi-Ying Yun
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (C.-Y.Y.); (T.Y.W.L.)
| | - Yao Neng Teo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
| | - Yao Hao Teo
- Internal Medicine Residency, National University Health System, Singapore 119074, Singapore; (E.H.E.T.); (Y.H.T.); (A.S.T.L.)
| | - Aloysius S. T. Leow
- Internal Medicine Residency, National University Health System, Singapore 119074, Singapore; (E.H.E.T.); (Y.H.T.); (A.S.T.L.)
| | - Tony Y. W. Li
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (C.-Y.Y.); (T.Y.W.L.)
| | - Vijay K. Sharma
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Division of Neurology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Benjamin Y. Q. Tan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Division of Neurology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Leonard L. L. Yeo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Division of Neurology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Yao Feng Chong
- Division of Neurology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Mark Y. Chan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (C.-Y.Y.); (T.Y.W.L.)
| | - Ching-Hui Sia
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (C.-Y.Y.); (T.Y.W.L.)
| |
Collapse
|
2
|
Statins in High Cardiovascular Risk Patients: Do Comorbidities and Characteristics Matter? Int J Mol Sci 2022; 23:ijms23169326. [PMID: 36012589 PMCID: PMC9409457 DOI: 10.3390/ijms23169326] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) morbidity and mortality are decreasing in high-income countries, but ASCVD remains the leading cause of morbidity and mortality in high-income countries. Over the past few decades, major risk factors for ASCVD, including LDL cholesterol (LDL-C), have been identified. Statins are the drug of choice for patients at increased risk of ASCVD and remain one of the most commonly used and effective drugs for reducing LDL cholesterol and the risk of mortality and coronary artery disease in high-risk groups. Unfortunately, doctors tend to under-prescribe or under-dose these drugs, mostly out of fear of side effects. The latest guidelines emphasize that treatment intensity should increase with increasing cardiovascular risk and that the decision to initiate intervention remains a matter of individual consideration and shared decision-making. The purpose of this review was to analyze the indications for initiation or continuation of statin therapy in different categories of patient with high cardiovascular risk, considering their complexity and comorbidities in order to personalize treatment.
Collapse
|
3
|
Sarzani R, Allevi M, Giulietti F, Di Pentima C, Re S, Giordano P, Spannella F. The Identikit of Patient at Risk for Severe COVID-19 and Death: The Dysregulation of Renin-Angiotensin System as the Common Theme. J Clin Med 2021; 10:5883. [PMID: 34945176 PMCID: PMC8704645 DOI: 10.3390/jcm10245883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 12/24/2022] Open
Abstract
Since the first months of the coronavirus disease 2019 (COVID-19) pandemic, several specific physiologic traits, such as male sex and older age, or health conditions, such as overweight/obesity, arterial hypertension, metabolic syndrome, and type 2 diabetes mellitus, have been found to be highly prevalent and associated with increased risk of adverse outcomes in hospitalized patients. All these cardiovascular morbidities are widespread in the population and often coexist, thus identifying a common patient phenotype, characterized by a hyper-activation of the "classic" renin-angiotensin system (RAS) and mediated by the binding of angiotensin II (Ang II) to the type 1-receptor. At the same time, the RAS imbalance was proved to be crucial in the genesis of lung injury after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, where angiotensin-converting-enzyme-2 (ACE2) is not only the receptor for SARS-CoV-2, but its down-regulation through internalization and shedding, caused by the virus binding, leads to a further dysregulation of RAS by reducing angiotensin 1-7 (Ang 1-7) production. This focused narrative review will discuss the main available evidence on the role played by cardiovascular and metabolic conditions in severe COVID-19, providing a possible pathophysiological link based on the disequilibrium between the two opposite arms of RAS.
Collapse
Affiliation(s)
- Riccardo Sarzani
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, Via Tronto 10/a, 60126 Ancona, Italy
| | - Massimiliano Allevi
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, Via Tronto 10/a, 60126 Ancona, Italy
| | - Federico Giulietti
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, Via Tronto 10/a, 60126 Ancona, Italy
| | - Chiara Di Pentima
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, Via Tronto 10/a, 60126 Ancona, Italy
| | - Serena Re
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, Via Tronto 10/a, 60126 Ancona, Italy
| | - Piero Giordano
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
| | - Francesco Spannella
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy; (M.A.); (F.G.); (C.D.P.); (S.R.); (P.G.); (F.S.)
- Department of Clinical and Molecular Sciences, University “Politecnica delle Marche”, Via Tronto 10/a, 60126 Ancona, Italy
| |
Collapse
|
4
|
Statins, toxicity, and their adverse effects via oxidative imbalance. Toxicology 2021. [DOI: 10.1016/b978-0-12-819092-0.00026-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
5
|
Dietl A, Maack C. Targeting Mitochondrial Calcium Handling and Reactive Oxygen Species in Heart Failure. Curr Heart Fail Rep 2017; 14:338-349. [PMID: 28656516 DOI: 10.1007/s11897-017-0347-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW In highly prevalent cardiac diseases, new therapeutic approaches are needed. Since the first description of oxidative stress in heart failure, reactive oxygen species (ROS) have been considered as attractive drug targets. Though clinical trials evaluating antioxidant vitamins as ROS-scavenging agents yielded neutral results in patients at cardiovascular risk, the knowledge of ROS as pathophysiological factors has considerably advanced in the past few years and led to novel treatment approaches. Here, we review recent new insights and current strategies in targeting mitochondrial calcium handling and ROS in heart failure. RECENT FINDINGS Mitochondria are an important ROS source, and more recently, drug development focused on targeting mitochondria (e.g. by SS-31 or MitoQ). Important advancement has also been made to decipher how the matching of energy supply and demand through calcium (Ca2+) handling impacts on mitochondrial ROS production and elimination. This opens novel opportunities to ameliorate mitochondrial dysfunction in heart failure by targeting cytosolic and mitochondrial ion transporters to improve this matching process. According to this approach, highly specific substances as the preclinical CGP-37157, as well as the clinically used ranolazine and empagliflozin, provide promising results on different levels of evidence. Furthermore, the understanding of redox signalling relays, resembled by catalyst-mediated protein oxidation, is about to change former paradigms of ROS signalling. Novel methods, as redox proteomics, allow to precisely analyse key regulatory thiol switches, which may induce adaptive or maladaptive signalling. Additionally, the generation of genetically encoded probes increased the spatial and temporal resolution of ROS imaging and opened a new methodological window to subtle, formerly obscured processes. These novel insights may broaden our understanding of why previous attempts to target oxidative stress have failed, and at the same time provide us with new targets for drug development.
Collapse
Affiliation(s)
- Alexander Dietl
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421, Homburg, Germany
| | - Christoph Maack
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421, Homburg, Germany.
| |
Collapse
|
6
|
D'Elia JA, Bayliss G, Gleason RE, Weinrauch LA. Cardiovascular-renal complications and the possible role of plasminogen activator inhibitor: a review. Clin Kidney J 2016; 9:705-12. [PMID: 27679717 PMCID: PMC5036907 DOI: 10.1093/ckj/sfw080] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 07/20/2016] [Indexed: 12/14/2022] Open
Abstract
Since angiotensin increases the expression of plasminogen activator inhibitor (PAI), mechanisms associated with an actively functioning renin–angiotensin–aldosterone system can be expected to be associated with increased PAI-1 expression. These mechanisms are present not only in common conditions resulting in glomerulosclerosis associated with aging, diabetes or genetic mutations, but also in autoimmune disease (like scleroderma and lupus), radiation injury, cyclosporine toxicity, allograft nephropathy and ureteral obstruction. While the renin–angiotensin–aldosterone system and growth factors, such as transforming growth factor-beta (TGF-β), are almost always part of the process, there are rare experimental observations of PAI-1 expression without their interaction. Here we review the literature on PAI-1 and its role in vascular, fibrotic and oxidative injury as well as work suggesting potential areas of intervention in the pathogenesis of multiple disorders.
Collapse
Affiliation(s)
- John A D'Elia
- Joslin Diabetes Center, Boston, MA, USA; Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - George Bayliss
- Division ofKidney Diseases and Hypertension, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA; The Miriam Hospital, Providence, RI, USA; Alpert Medical School, Brown University, Providence, RI, USA
| | - Ray E Gleason
- Joslin Diabetes Center, Boston, MA, USA; Beth Israel Deaconess Medical Center, Boston, MA, USA; EP Joslin Research Laboratory, Boston, MA, USA; Brigham and Women's Hospital, Boston, MA, USA
| | - Larry A Weinrauch
- Joslin Diabetes Center, Boston, MA, USA; Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; EP Joslin Research Laboratory, Boston, MA, USA; Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
7
|
Atorvastatin improves cardiac function and remodeling in chronic non-ischemic heart failure: A clinical and pre-clinical study. Egypt Heart J 2015. [DOI: 10.1016/j.ehj.2014.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
8
|
Al-Rasheed NM, Al-Oteibi MM, Al-Manee RZ, Al-Shareef SA, Al-Rasheed NM, Hasan IH, Mohamad RA, Mahmoud AM. Simvastatin prevents isoproterenol-induced cardiac hypertrophy through modulation of the JAK/STAT pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3217-29. [PMID: 26150695 PMCID: PMC4484667 DOI: 10.2147/dddt.s86431] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Simvastatin (SIM) is a lipid-soluble inhibitor of hydroxy-3-methylglutaryl coenzyme A reductase with multiple reported therapeutic benefits. The present study was designed to investigate the effect of pretreatment with SIM on isoproterenol (ISO)-induced cardiac hypertrophy in rats. Twenty-four male albino Wistar rats weighing 180–200 g were divided into four groups. Groups I and III received normal saline while groups II and IV received SIM (10 mg/kg body weight) for 30 days per gavage. In the last 7 days, rats of groups III and IV were administered ISO (5 mg/kg) intraperitoneally to induce cardiac hypertrophy. Administration of ISO induced an increase in heart-to-body weight (HW/BW) ratio, an increase in serum interleukin-6, and elevated systolic and diastolic blood pressure. Serum levels of lipids, cardiovascular risk indices, and cardiac troponin I and creatine phosphokinase-MB showed significant increase in ISO-induced hypertrophic rats. Histopathological examination of heart tissue revealed focal areas of subendocardium degeneration, mononuclear cellular infiltrations, fibrous tissue deposition, and increased thickness of the myocardium of left ventricle. In addition, ISO-administered rats exhibited significant upregulation of cardiac Janus kinase, phosphorylated signal transducer and activator of transcription, and nuclear factor-kappa B. Pretreatment with SIM significantly prevented ISO-induced cardiac hypertrophy, alleviated the altered biochemical parameters, and improved the heart architecture. In conclusion, our study provides evidence that SIM prevented the development of cardiac hypertrophy via modulation of the Janus kinase/signal transducer and activator of transcription-signaling pathway in the heart of ISO-administered animals.
Collapse
Affiliation(s)
- Nouf M Al-Rasheed
- Department of Pharmacology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Maha M Al-Oteibi
- Department of Pharmacology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Reem Z Al-Manee
- Department of Pharmacology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sarah A Al-Shareef
- Department of Pharmacology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nawal M Al-Rasheed
- Department of Pharmacology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Iman H Hasan
- Department of Pharmacology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Raeesa A Mohamad
- Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ayman M Mahmoud
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Egypt
| |
Collapse
|
9
|
Hu HJ, Zhou SH, Liu QM. Treatment of pheochromocytoma blockade of MAPK pathway inhibition in the NF-κB pathway and bFGF — Effect of statins on pheochromocytoma patients. Int J Cardiol 2015; 182:161-2. [DOI: 10.1016/j.ijcard.2015.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 01/02/2015] [Indexed: 02/06/2023]
|
10
|
Ruggieri A, Gambardella L, Maselli A, Vona R, Anticoli S, Panusa A, Malorni W, Matarrese P. Statin-induced impairment of monocyte migration is gender-related. J Cell Physiol 2014; 229:1990-8. [PMID: 24777636 DOI: 10.1002/jcp.24657] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/25/2014] [Indexed: 11/07/2022]
Abstract
Statins, widely used for treatment of hypercholesterolemia, have been demonstrated to exert pleiotropic beneficial effects independently of their cholesterol-lowering action, such as anti-inflammatory activity. A gender disparity has been observed in their cholesterol lowering activity as well as in response to these "off label" effects. Monocytes play a central role in atherosclerotic disease and, more in general, in inflammatory responses, through their chemotactic function and cytokine production. On these bases, in the present work, we examined the effect of statins on homeostasis and migration properties of freshly isolated monocytes from male and female healthy donors. Two prototypic natural and synthetic statins with different polarity, that is, type 1 and type 2 statins, have been considered: simvastatin and atorvastatin. Freshly isolated monocytes from peripheral blood of male and female healthy donors were treated with these drugs in the absence or presence of lipopolysaccharide (LPS) stimulation. Results obtained indicated that the polar statin efficiently inhibited chemotaxis of monocytes more than the apolar statin and that this effect was more significantly induced in cells from females than in cells from males. Dissecting the mechanisms involved, we found that these results could mainly be due to differential effects on: (i) the release of key cytokines, for example, MCP-1 and TNF-α; (ii) the maintenance of the redox homeostasis; (iii) a target activity on microfilament network integrity and function. All in all these results could suggest a reappraisal of "off-label" effects of statins taking into account either their chemical structure, that is, molecular polarity, or the gender issue.
Collapse
Affiliation(s)
- Anna Ruggieri
- Department of Infectious, Parasitic and Immune Mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Markó L, Henke N, Park JK, Spallek B, Qadri F, Balogh A, Apel IJ, Oravecz-Wilson KI, Choi M, Przybyl L, Binger KJ, Haase N, Wilck N, Heuser A, Fokuhl V, Ruland J, Lucas PC, McAllister-Lucas LM, Luft FC, Dechend R, Müller DN. Bcl10 mediates angiotensin II-induced cardiac damage and electrical remodeling. Hypertension 2014; 64:1032-9. [PMID: 25185127 DOI: 10.1161/hypertensionaha.114.03900] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Angiotensin (Ang) II is a potent mediator of both hypertension and cardiac damage; however, the mechanisms by which this occur remain unclear. B-cell lymphoma/leukemia 10 (Bcl10) is a member of the CBM signalosome, which links Ang II and nuclear factor-κB signaling. We hypothesized that Bcl10 is pivotal in the pathogenesis of Ang II-induced cardiac damage. Ang II infusion in mice lacking Bcl10 resulted in reduced cardiac fibrosis, less cellular infiltration, and improved arrhythmogenic electric remodeling, despite a similar degree of hypertension or cardiac hypertrophy. Adoptive transfer of bone marrow (BM), whereby Bcl10 knockout or wildtype BM was transferred to their opposite genotype recipients, revealed the dual importance of Bcl10 within both cardiac and immune cells. Loss of Bcl10 in cardiac cells resulted in reduced expression of genes important for the adhesion and recruitment of immune cells. In vitro experiments demonstrated that adhesion of monocytes to Ang II-treated endothelial cells also required Bcl10. Additionally, Bcl10 deficiency in macrophages reduced their intrinsic migratory ability. To address the role of BM-derived fibroblasts in the formation of cardiac fibrosis, we explored whether Bcl10 is also important for the infiltration of BM-derived (myo)fibroblasts into the heart. The transfer of green fluorescent protein positive wildtype BM into Bcl10 knockout recipient mice revealed a reduced number of noncardiac (myo)fibroblasts compared with those wildtype recipients. Our results demonstrate the significant role of Bcl10 in multiple cell types important for the generation of Ang II-induced cardiac damage and electric remodeling and may provide a new avenue for therapeutic intervention.
Collapse
Affiliation(s)
- Lajos Markó
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Norbert Henke
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Joon-Keun Park
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Bastian Spallek
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Fatimunnisa Qadri
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - András Balogh
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Ingrid J Apel
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Katherine I Oravecz-Wilson
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Mira Choi
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Lukasz Przybyl
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Katrina J Binger
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Nadine Haase
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Nicola Wilck
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Arnd Heuser
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Verena Fokuhl
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Jürgen Ruland
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Peter C Lucas
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Linda M McAllister-Lucas
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Friedrich C Luft
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Ralf Dechend
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.)
| | - Dominik N Müller
- From the Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., B.S., F.Q., A.B., M.C., L.P., K.J.B., N.H., N.W., V.F., F.C.L., R.D., D.N.M.); Department of Internal Medicine/Cardiology, Helios Clinic Damp, Damp, Germany (N.H.); Clinic for Nephrology and Hypertension, Hannover Medical School, Hannover, Germany (J.K.P.); Department of Pathology, University of Michigan Medical School, Ann Arbor, MI (I.J.A., K.I.O.W.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (L.M., A.B., K.J.B., A.H., F.C.L., D.N.M.); Institute for Clinical Chemistry and Biochemistry, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany (J.R.); Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA (P.C.L.); Department of Pediatrics, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh, Pittsburgh, PA (L.M.M.-L.); and Department of Cardiology and Nephrology, Helios Clinic Berlin-Buch, Berlin, Germany (R.D.).
| |
Collapse
|
12
|
Pecoraro V, Moja L, Dall'Olmo L, Cappellini G, Garattini S. Most appropriate animal models to study the efficacy of statins: a systematic review. Eur J Clin Invest 2014; 44:848-71. [PMID: 25066257 DOI: 10.1111/eci.12304] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 07/21/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND In animal models and clinical trials, statins are reported as effective in reducing cholesterol levels and lowering the risk of cardiovascular diseases. We have aggregated the findings in animal models - mice, rats and rabbits - using the technique of systematic review and meta-analysis to highlight differences in the efficacy of statins. MATERIALS AND METHODS We searched Medline and Embase. After examining all eligible articles, we extracted results about total cholesterol and other blood parameters, blood pressure, myocardial infarction and survival. Weighted and standard mean difference random effects meta-analysis was used to measure overall efficacy in prespecified species, strains and subgroups. RESULTS We included in systematic review 161 animal studies and we analysed 120 studies, accounting for 2432 animals. Statins lowered the total cholesterol across all species, although with large differences in the effect size: -30% in rabbits, -20% in mice and -10% in rats. The reduction was larger in animals fed on a high-cholesterol diet. Statins reduced infarct volume but did not consistently reduce the blood pressure or effect the overall survival. Few studies considered strains at high risk of cardiovascular diseases or hard outcomes. CONCLUSIONS Although statins showed substantial efficacy in animal models, few preclinical data considered conditions mimicking human pathologies for which the drugs are clinically indicated and utilized. The empirical finding that statins are more effective in lowering cholesterol derived from an external source (i.e. diet) conflicts with statin's supposed primary mechanism of action.
Collapse
Affiliation(s)
- Valentina Pecoraro
- Clinical Epidemiology Unit, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | | | | | | | | |
Collapse
|
13
|
Haase N, Rugor J, Przybyl L, Qadri F, Müller DN, Dechend R. Relaxin does not improve Angiotensin II-induced target-organ damage. PLoS One 2014; 9:e93743. [PMID: 24710077 PMCID: PMC3977876 DOI: 10.1371/journal.pone.0093743] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 03/06/2014] [Indexed: 11/23/2022] Open
Abstract
Relaxin is a corpus-luteum produced protein hormone with vasodilatatory, anti-fibrotic, and angiogenic properties that are opposite to angiotensin (Ang) II. We investigated whether or not relaxin ameliorates Ang II-induced target-organ damage. We used double transgenic rats harboring both human renin and angiotensinogen genes (dTGR) that develop severe hypertension, target-organ damage, and die untreated within 7–8 weeks. Recombinant relaxin at a low (26 μg/kg/d) and a high dose (240 μg/kg/d) was given to 4 week-old dTGR and age-matched Sprague-Dawley rats (SD). Systolic blood pressure increased progressively in untreated dTGRs from 162±3 mmHg at week 5 to 225±5 mmHg at week 7. Relaxin had no effect on blood pressure whereas SD rats were normotensive (106±1 mmHg). Untreated and relaxin-treated dTGR had similarly severe cardiac hypertrophy indices. Relaxin did not ameliorate albuminuria and did not prevent matrix-protein deposition in the heart and kidney in dTGR. Finally, relaxin treatment did not reduce mortality. These data suggest that pharmacological doses of relaxin do not reverse severe effects of Ang II.
Collapse
Affiliation(s)
- Nadine Haase
- Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrueck Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany
| | - Julianna Rugor
- Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrueck Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany
| | - Lukasz Przybyl
- Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrueck Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany
| | - Fatimunnisa Qadri
- Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrueck Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany
| | - Dominik N. Müller
- Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrueck Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrueck Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany
- Department of Cardiology and Nephrology, HELIOS-Klinikum Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
14
|
Margaritis M, Channon KM, Antoniades C. Statins as regulators of redox state in the vascular endothelium: beyond lipid lowering. Antioxid Redox Signal 2014; 20:1198-215. [PMID: 24111702 PMCID: PMC3934595 DOI: 10.1089/ars.2013.5430] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Endothelial dysfunction and the imbalance between nitric oxide (NO) and reactive oxygen species production in the vascular endothelium are important early steps in atherogenesis, a major socioeconomic health problem. Statins have well-established roles in primary and secondary prevention of cardiovascular disease (CVD), due to both their lipid-lowering capacity and their pleiotropic properties. It is therefore important to understand the mechanisms by which statins can modify endothelial function and affect atherogenesis. RECENT ADVANCES In the last decade, the concept of statin pleiotropy has been reinforced by a large number of cell culture, animal, and translational studies. Statins have been shown to suppress the activity of pro-oxidant enzymes (such as NADPH oxidase) and pro-inflammatory transcriptional pathways in the endothelium. At the same time, they enhance endothelial NO synthase expression and activity while they also improve its enzymatic coupling. This leads to increased NO bioavailability and improved endothelial function. CRITICAL ISSUES Despite significant recent advances, the exact mechanisms of statin pleitropy are still only partially understood. The vast majority of the published literature relies on animal studies, while the actual mechanistic studies in humans are limited. FUTURE DIRECTIONS The success of statins as endothelium redox-modifying agents with a direct impact on clinical outcome highlights the importance of the endothelium as a therapeutic target in CVD. Better understanding of the mechanisms that underlie endothelial dysfunction could lead to the design of novel therapeutic strategies that target the vascular endothelium for the prevention and treatment of CVD.
Collapse
Affiliation(s)
- Marios Margaritis
- Division of Cardiovascular Medicine, University of Oxford , Oxford, United Kingdom
| | | | | |
Collapse
|
15
|
Adam O, Laufs U. Rac1-mediated effects of HMG-CoA reductase inhibitors (statins) in cardiovascular disease. Antioxid Redox Signal 2014; 20:1238-50. [PMID: 23919665 DOI: 10.1089/ars.2013.5526] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE HMG-CoA reductase inhibitors (statins) lower serum cholesterol concentrations and are beneficial in the primary and secondary prevention of coronary heart disease. The positive clinical effects have only partially been reproduced with other lipid-lowering interventions suggesting potential statin effects in addition to cholesterol lowering. In experimental models, direct beneficial cardiovascular effects that are mediated by the inhibition of isoprenoids have been documented, which serve as lipid attachments for intracellular signaling molecules such as small Rho guanosine triphosphate-binding proteins, whose membrane localization and function are dependent on isoprenylation. RECENT ADVANCES Rac1 GTPase is an established master regulator of cell motility through the cortical actin reorganization and of reactive oxygen species generation through the regulation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity. CRITICAL ISSUES Observations in cells, animals, and humans have implicated the activation of Rac1 GTPase as a key component of cardiovascular pathologies, including the endothelial dysfunction, cardiac hypertrophy and fibrosis, atrial fibrillation, stroke, hypertension, and chronic kidney disease. However, the underlying signal transduction remains incompletely understood. FUTURE DIRECTIONS Based on the recent advance made in Rac1 research in the cardiovascular system by using mouse models with transgenic overexpression of activated Rac1 or conditional knockout, as well as Rac1-specific small molecule inhibitor NSC 23766, the improved understanding of the Rac1-mediated effects statins may help to identify novel therapeutic targets and strategies.
Collapse
Affiliation(s)
- Oliver Adam
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes , Homburg, Germany
| | | |
Collapse
|
16
|
Yeganeh B, Wiechec E, Ande SR, Sharma P, Moghadam AR, Post M, Freed DH, Hashemi M, Shojaei S, Zeki AA, Ghavami S. Targeting the mevalonate cascade as a new therapeutic approach in heart disease, cancer and pulmonary disease. Pharmacol Ther 2014; 143:87-110. [PMID: 24582968 DOI: 10.1016/j.pharmthera.2014.02.007] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/04/2014] [Indexed: 12/21/2022]
Abstract
The cholesterol biosynthesis pathway, also known as the mevalonate (MVA) pathway, is an essential cellular pathway that is involved in diverse cell functions. The enzyme 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase (HMGCR) is the rate-limiting step in cholesterol biosynthesis and catalyzes the conversion of HMG-CoA to MVA. Given its role in cholesterol and isoprenoid biosynthesis, the regulation of HMGCR has been intensely investigated. Because all cells require a steady supply of MVA, both the sterol (i.e. cholesterol) and non-sterol (i.e. isoprenoid) products of MVA metabolism exert coordinated feedback regulation on HMGCR through different mechanisms. The proper functioning of HMGCR as the proximal enzyme in the MVA pathway is essential under both normal physiologic conditions and in many diseases given its role in cell cycle pathways and cell proliferation, cholesterol biosynthesis and metabolism, cell cytoskeletal dynamics and stability, cell membrane structure and fluidity, mitochondrial function, proliferation, and cell fate. The blockbuster statin drugs ('statins') directly bind to and inhibit HMGCR, and their use for the past thirty years has revolutionized the treatment of hypercholesterolemia and cardiovascular diseases, in particular coronary heart disease. Initially thought to exert their effects through cholesterol reduction, recent evidence indicates that statins also have pleiotropic immunomodulatory properties independent of cholesterol lowering. In this review we will focus on the therapeutic applications and mechanisms involved in the MVA cascade including Rho GTPase and Rho kinase (ROCK) signaling, statin inhibition of HMGCR, geranylgeranyltransferase (GGTase) inhibition, and farnesyltransferase (FTase) inhibition in cardiovascular disease, pulmonary diseases (e.g. asthma and chronic obstructive pulmonary disease (COPD)), and cancer.
Collapse
Affiliation(s)
- Behzad Yeganeh
- Hospital for Sick Children Research Institute, Department of Physiology & Experimental Medicine, University of Toronto, Toronto, Canada
| | - Emilia Wiechec
- Dept. Clinical & Experimental Medicine, Division of Cell Biology & Integrative Regenerative Med. Center (IGEN), Linköping University, Sweden
| | - Sudharsana R Ande
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pawan Sharma
- Department of Physiology & Pharmacology, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, 4C46 HRIC, 3280 Hospital Drive NW, Calgary, Alberta, Canada
| | - Adel Rezaei Moghadam
- Scientific Association of Veterinary Medicine, Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran; Young Researchers and Elite Club, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| | - Martin Post
- Hospital for Sick Children Research Institute, Department of Physiology & Experimental Medicine, University of Toronto, Toronto, Canada
| | - Darren H Freed
- Department of Physiology, St. Boniface Research Centre, University of Manitoba, Winnipeg, Canada
| | - Mohammad Hashemi
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Shahla Shojaei
- Department of Biochemistry, Recombinant Protein Laboratory, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir A Zeki
- U.C. Davis, School of Medicine, U.C. Davis Medical Center, Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Center for Comparative Respiratory Biology & Medicine, Davis, CA, USA.
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, St. Boniface Research Centre, Manitoba Institute of Child Health, Biology of Breathing Theme, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
17
|
Chen Y, Teng X, Chen W, Yang J, Yang Z, Yu Y, Shen Z. Timing of transplantation of autologous bone marrow derived mesenchymal stem cells for treating myocardial infarction. SCIENCE CHINA-LIFE SCIENCES 2014; 57:195-200. [DOI: 10.1007/s11427-013-4605-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 10/02/2013] [Indexed: 11/24/2022]
|
18
|
Marchioli R, Levantesi G, Silletta MG, Barlera S, Bernardinangeli M, Carbonieri E, Cosmi F, Franzosi MG, Latini R, Lucci D, Maggioni AP, Moretti L, Nicolosi GL, Porcu M, Rossi MG, Tognoni G, Tavazzi L. Effect of n-3 polyunsaturated fatty acids and rosuvastatin in patients with heart failure: results of the GISSI-HF trial. Expert Rev Cardiovasc Ther 2014; 7:735-48. [DOI: 10.1586/erc.09.70] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
Statin-Mediated Low-Density Lipoprotein Lowering in Chronic Congestive Heart Failure. Am J Med Sci 2014; 347:14-22. [DOI: 10.1097/maj.0b013e318273514c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
20
|
Abstract
Statins lower serum cholesterol and are employed for primary and secondary prevention of cardiovascular events. Clinical evidence from observational studies, retrospective data, and post hoc analyses of data from large statin trials in various cardiovascular conditions, as well as small scale randomized trials, suggest survival and other outcome benefits for heart failure. Two recent large randomized controlled trials, however, appear to suggest statins do not have beneficial effects in heart failure. In addition to lowering cholesterol, statins are believed to have many pleotropic effects which could possibly influence the pathophysiology of heart failure. Following the two large trials, evidence from recent studies appears to support the use of statins in heart failure. This review discusses the role of statins in the pathophysiology of heart failure, current evidence for statin use in heart failure, and suggests directions for future research.
Collapse
Affiliation(s)
- Kwadwo Osei Bonsu
- School of Medicine and Health Sciences, Monash University Sunway Campus, Bandar Sunway, Malaysia
| | | | | |
Collapse
|
21
|
Abstract
Hydroxy methyl glutaryl CoA inhibitors (statins) are the agents most frequently used to reduce elevated serum cholesterol. In addition to their cholesterol lowering effects, statins also have nonlipid lowering pleiotropic properties. These include reducing oxidative stress, renin-angiotensin and endothelin synthesis and activity, and improving nitric oxide (NO) synthesis and availability. Thus, one would predict that statins might be able to exert an antihypertensive effect. Experimental models bear out the blood pressure lowering effects but the data from clinical trials have been inconsistent perhaps due to inappropriate experimental designs, sample size, blood pressure measurement techniques etc. Moreover, although experimental models strongly suggest a role for salt intake in the potential antihypertensive responses to statins, available clinical trials fail to report salt intake in the studied populations. The statins' antihypertensive effects remain an unsettled hypothesis and calls for a large clinical trial at a wide range of doses and a controlled salt intake. Statins meanwhile remain as a excellent option to control high cholesterol and in tissue injury prevention.
Collapse
|
22
|
Norata GD. Established and emerging approaches for the management of dyslipidaemia. SCIENTIFICA 2012; 2012:482423. [PMID: 24278703 PMCID: PMC3820450 DOI: 10.6064/2012/482423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 08/26/2012] [Indexed: 06/02/2023]
Abstract
The key role of dyslipidaemia in determining cardiovascular disease (CVD) has been proved beyond reasonable doubt, and therefore several dietary and pharmacological approaches have been developed. The discovery of statins has provided a very effective approach in reducing cardiovascular risk as documented by the results obtained in clinical trials and in clinical practice. The current efficacy of statins or other drugs, however, comes short of providing the benefit that could derive from a further reduction of LDL cholesterol (LDL-C) in high-risk and very high risk patients. Furthermore, experimental data clearly suggest that other lipoprotein classes beyond LDL play important roles in determining cardiovascular risk. For these reasons a number of new potential drugs are under development in this area. Aim of this review is to discuss the available and the future pharmacological strategies for the management of dyslipidemia.
Collapse
Affiliation(s)
- Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano 20122 Milan, Italy
- Center for the Study of Atherosclerosis, Società Italiana Studio Aterosclerosi, Ospedale Bassini, 20092 Cinisello Balsamo, Italy
- Centre for Diabetes, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University, London E12AT, UK
| |
Collapse
|
23
|
|
24
|
Copaja M, Venegas D, Aranguiz P, Canales J, Vivar R, Avalos Y, Garcia L, Chiong M, Olmedo I, Catalán M, Leyton L, Lavandero S, Díaz-Araya G. Simvastatin disrupts cytoskeleton and decreases cardiac fibroblast adhesion, migration and viability. Toxicology 2012; 294:42-9. [PMID: 22306966 DOI: 10.1016/j.tox.2012.01.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 01/16/2012] [Accepted: 01/20/2012] [Indexed: 12/31/2022]
Abstract
Statins reduce the isoprenoids farnesyl and geranylgeranyl pyrophosphate, essential intermediates, which control a diversity of cellular events such as cytoskeleton integrity, adhesion, migration and viability. Cardiac fibroblasts are the major non-myocyte cell constituent in the normal heart, and play a key role in the maintenance of extracellular matrix. The effects of simvastatin on cardiac fibroblast processes previously mentioned remain unknown. Our aims were to investigate the effects of simvastatin on cytoskeleton structure and focal adhesion complex assembly and their relationships with cell adhesion, migration and viability in cultured cardiac fibroblasts. To this end, cells were treated with simvastatin for 24 h and changes in actin cytoskeleton, levels of vimentin and paxillin as well as their subcellular localization were analyzed by Western blot and immunocytochemistry, respectively. Cell adhesion to plastic or collagen coated dishes, migration in Transwell chambers, and cell viability were analyzed after simvastatin treatment. Our results show that simvastatin disrupts actin cytoskeleton and focal adhesion complex evaluated by phalloidin stain and immunocytochemistry for paxillin and vinculin. All these effects occurred by a cholesterol synthesis-independent mechanism. Simvastatin decreased cell adhesion, migration and viability in a concentration-dependent manner. Finally, simvastatin decreased angiotensin II-induced phospho-paxillin levels and cell adhesion. We concluded that simvastatin disrupts cytoskeleton integrity and focal adhesion complex assembly in cultured cardiac fibroblasts by a cholesterol-independent mechanism and consequently decreases cell migration, adhesion and viability.
Collapse
Affiliation(s)
- Miguel Copaja
- Centro Estudios Moleculares de la Célula, Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Fassett RG, Robertson IK, Ball MJ, Geraghty DP, Cardinal JW, Coombes JS. Effects of atorvastatin on NGAL and cystatin C in chronic kidney disease: a post hoc analysis of the LORD trial. Nephrol Dial Transplant 2012; 27:182-189. [DOI: 10.1093/ndt/gfr193] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
26
|
Zhang J, Xu Y, Pan L, Chen T, Chen Z, Zhao R. Effect of simvastatin on collagen I deposition in non-infarcted myocardium: role of NF-κB and osteopontin. Can J Physiol Pharmacol 2011; 88:1026-34. [PMID: 21076490 DOI: 10.1139/y10-075] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The novel biological effect of statins in alleviating myocardium fibrosis following infarction has been increasingly recognized, yet the underlying mechanisms are not fully understood. The purpose of this study was to characterize the effect of simvastatin on myocardial fibrosis and collagen I deposition in the non-infarcted region after myocardial infarction (MI) and to identify the role of NF-κB and osteopontin in simvastatin-mediated inhibition of post-MI collagen over-expression. A rat model of MI was generated by ligating the left anterior descending coronary artery. The rats surviving the MI operation were randomly divided into the following 3 groups: myocardial infarction (MI, vehicle), simvastatin (Sim, 30 mg·kg-1·day-1), and pyrrolidine dithiocarbamate (PDTC, an inhibitor of NF-κB, 100 mg·kg-1·day-1). Four weeks after MI, cardiac function, mRNAs, and protein expression in non-infarcted myocardium were analyzed. Myocardial fibrosis and collagen I over-expression were observed following MI, accompanied by an increase of NF-κB and osteopontin. Simvastatin improved post-MI left ventricular dysfunction and ameliorated post-MI associated changes to several cardiac parameters, including the left ventricular end diastolic pressure (LVEDP), the maximal rate of pressure development (+dP/dtmax), and the maximal rate of pressure decline (-dP/dtmax). Concurrently, simvastatin significantly suppressed the over-expression of NF-κB, osteopontin, and collagen I in the non-infarcted region following MI. Inhibition of NF-κB by PDTC also reduced osteopontin over-expression and excessive collagen I production and improved the above functional myocardial parameters. These results show that post-MI myocardial fibrosis and collagen I over-expression in the non-infarcted region is associated with activation of NF-κB and osteopontin up-regulation. The anti-fibrotic effect of simvastatin following MI is associated with the attenuation of the expression of osteopontin and NF-κB. The inhibition of NF-κB activation could be the process upstream of osteopontin suppression in the simvastatin-mediated effect.
Collapse
Affiliation(s)
- Jianhua Zhang
- Department of Cardiology, First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China
| | | | | | | | | | | |
Collapse
|
27
|
Inanc MT, Kalay N, Heyit T, Ozdogru I, Kaya MG, Dogan A, Duran M, Kasapkara HA, Gunebakmaz O, Borlu M, Yarlıoglues M, Oguzhan A. Effects of atorvastatin and lisinopril on endothelial dysfunction in patients with Behçet's disease. Echocardiography 2011; 27:997-1003. [PMID: 20545993 DOI: 10.1111/j.1540-8175.2010.01180.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE Behçet's disease is a chronic inflammatory vasculitis. Vascular involvement is one of the major complications of Behçet's disease, during the course of the disease. Previous studies showed that ACE inhibitors and statins may improve endothelial functions in endothelial dysfunction. The aim of our study is to compare the effects of atorvastatin and lisinopril to placebo on endothelial dysfunction in patients with Behçet's disease. PATIENTS AND METHODS We prospectively studied 92 (48 female) Behçet's patients who were diagnosed according to the International Study Group criteria. Endothelial dysfunction was evaluated by brachial artery flow-mediated dilatation (FMD) method using high-resolution vascular ultrasound device at baseline and after for 3-month therapy. Patients were consecutively randomized into three groups as (atorvastatin (n = 31), lisinopril (n = 31), and placebo groups (n = 30). Patients in atorvastatin group received 20 mg atorvastatin, lisinopril group received 10 mg lisinopril per day, and placebo group received placebo per day for 3 months. RESULTS The baseline characteristics of patients were similar among three groups; however, high-sensitive C-reactive protein (hs-CRP) levels were lower in atorvastatin group than placebo group. A significant improvement in FMD was observed in both atorvastatin (5.0 ± 1.4 vs. 12.8 ± 3.6%, P < 0.001) and lisinopril groups (5.0 ± 1.2 vs. 11.4 ± 5.0%, P < 0.001). Partial significant enhancement was observed in placebo group (4.9 ± 1.1% vs. 5.7 ± 1.0, P = 0.002). However, it was lower than the cutoff value for endothelial dysfunction. CONCLUSION These findings suggest that atorvastatin and lisinopril improve endothelial functions in Behçet's disease patients. However, large studies are needed to determine the long-term effects of atorvastatin and lisinopril therapy.
Collapse
|
28
|
Preusch MR, Vanakaris A, Bea F, Ieronimakis N, Shimizu T, Konstandin M, Morris-Rosenfeld S, Albrecht C, Kranzhöfer A, Katus HA, Blessing E, Kranzhöfer R. Rosuvastatin reduces neointima formation in a rat model of balloon injury. Eur J Med Res 2011; 15:461-7. [PMID: 21159570 PMCID: PMC3352654 DOI: 10.1186/2047-783x-15-11-461] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Processes of restenosis, following arterial injury, are complex involving different cell types producing various cytokines and enzymes. Among those enzymes, smooth muscle cell-derived matrix metalloproteinases (MMPs) are thought to take part in cell migration, degrading of extracellular matrix, and neointima formation. MMP-9, also known as gelatinase B, is expressed immediately after vascular injury and its expression and activity can be inhibited by statins. Using an established in vivo model of vascular injury, we investigated the effect of the HMG-CoA reductase inhibitor rosuvastatin on MMP-9 expression and neointima formation. MATERIALS AND METHODS 14-week old male Sprague Dawley rats underwent balloon injury of the common carotid artery. Half of the animals received rosuvastatin (20 mg/kg body weight/day) via oral gavage, beginning 3 days prior to injury. Gelatinase activity and neointima formation were analyzed 3 days and 14 days after balloon injury, respectively. 14 days after vascular injury, proliferative activity was assessed by staining for Ki67. RESULTS After 14 days, animals in the rosuvastatin group showed a decrease in total neointima formation (0.194±0.01 mm2 versus 0.124±0.02 mm2, p<0.05) as well as a reduced intima/media ratio (1.26±0.1 versus 0.75±0.09, p<0.05). Balloon injury resulted in increased activity of MMP-9 3 days after intervention for both rosuvastatin treated animals and controls with no significant difference observed between the groups. There was a trend towards a reduction in the number of Ki67-positive cells 14 days after injury. CONCLUSIONS Rosuvastatin attenuates neointima formation without affecting early MMP-9 activity in a rat model of vascular injury.
Collapse
Affiliation(s)
- M R Preusch
- Department of Internal Medicine III, University of Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
No effect of rosuvastatin on left ventricular hypertrophy in patients with hypertension. Int J Cardiol 2010; 145:156-8. [DOI: 10.1016/j.ijcard.2009.07.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 07/25/2009] [Indexed: 11/22/2022]
|
30
|
Effects of fluvastatin on insulin resistance and cardiac morphology in hypertensive patients. J Hum Hypertens 2010; 25:492-9. [PMID: 20827284 DOI: 10.1038/jhh.2010.87] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Among hypertensive patients, cardiovascular disease morbidity is common, even in those who are adequately treated. New pharmacological strategies to mitigate the burden of arterial hypertension are needed. This 12-month, randomized, double-blind placebo-controlled study investigated the effect of statin (fluvastatin) treatment on ambulatory blood pressure (ABP), exercise blood pressure (EBP), myocardial structure, endothelial function and insulin resistance in 50 hypertensive patients. At baseline, the groups were comparable in terms of demographic characteristics, ABP, EBP, endothelial function and homeostasis model assessment of insulin resistance (HOMA-IR). At the end of the study, there was no difference between groups in terms of resting systolic blood pressure. However, maximum systolic EBP was lower in the treatment group than in the placebo group (175 ± 18 vs 192 ± 23 mm Hg, P<0.05), as was left ventricular mass index (LVMI; 82 ± 15 vs 100 ± 23, P<0.05), and HOMA-IR index was lower after fluvastatin treatment (2.77 ± 1.46 vs 3.33 ± 1.73, P<0.05). Changes in lipid profile were not correlated with blood pressure, endothelial function, LVMI or HOMA-IR data. In hypertensive patients, fluvastatin can improve maximum systolic EBP, myocardial remodelling and insulin resistance, independently of lipid profile variations and endothelial function.
Collapse
|
31
|
From aldosteronism to oxidative stress: the role of excessive intracellular calcium accumulation. Hypertens Res 2010; 33:1091-101. [DOI: 10.1038/hr.2010.159] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
32
|
Sadowitz B, Seymour K, Costanza MJ, Gahtan V. Basic Science Review Section: Statin Therapy—Part II: Clinical Considerations for Cardiovascular Disease. Vasc Endovascular Surg 2010; 44:421-33. [DOI: 10.1177/1538574410363833] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
3-Hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, commonly known as statins, are the medical treatment of choice for hypercholesterolemia. In addition to achieving a therapeutic decrease in serum cholesterol levels, statin therapy appears to promote pleiotropic effects that are independent of changes in serum cholesterol. These cholesterol lowering and pleiotropic effects are beneficial not only for the coronary circulation, but for the myocardium and peripheral arterial system as well. Patients receiving statin therapy must be carefully monitored, however, as statins potentially have harmful side effects and drug interactions. This article is part II of a 2-part review, and it focuses on the clinical aspects of statin therapy in cardiovascular disease.
Collapse
Affiliation(s)
- Benjamin Sadowitz
- Division of Vascular Surgery and Endovascular Services, SUNY Upstate Medical University, Syracuse, NY, USA, Department of Veterans Affairs, VA Healthcare Network Upstate New York at Syracuse, Syracuse, NY, USA
| | - Keri Seymour
- Division of Vascular Surgery and Endovascular Services, SUNY Upstate Medical University, Syracuse, NY, USA, Department of Veterans Affairs, VA Healthcare Network Upstate New York at Syracuse, Syracuse, NY, USA
| | - Michael J. Costanza
- Division of Vascular Surgery and Endovascular Services, SUNY Upstate Medical University, Syracuse, NY, USA, , Department of Veterans Affairs, VA Healthcare Network Upstate New York at Syracuse, Syracuse, NY, USA
| | - Vivian Gahtan
- Division of Vascular Surgery and Endovascular Services, SUNY Upstate Medical University, Syracuse, NY, USA, Department of Veterans Affairs, VA Healthcare Network Upstate New York at Syracuse, Syracuse, NY, USA
| |
Collapse
|
33
|
Gong C, Huang SL, Huang JF, Zhang ZF, Luo M, Zhao Y, Jiang XJ. Effects of combined therapy of Xuezhikang Capsule and Valsartan on hypertensive left ventricular hypertrophy and heart rate turbulence. Chin J Integr Med 2010; 16:114-8. [PMID: 20473735 DOI: 10.1007/s11655-010-0114-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To observe the effect of combined therapy with Xuezhikang Capsule (XZK) and Valsartan on left ventricular hypertrophy (LVH) and heart rate turbulence (HRT) in hypertensive patients. METHODS Ninety primary hypertensive patients with LVH were randomly assigned to three groups. Basic treatment, including aspirin, beta-blockers, calcium antagonists, etc. were administered to all patients. Additionally, Valsartan (VS, 80 mg once a day) was given to the 30 patients in the VS group. Valsartan (in the same dosage) and XZK (600 mg, twice a day) were given to the 32 patients in the Chinese medicine (CM) group, while none was given to the 28 patients in the control group. The therapeutic course lasted for 24 months. Changes in left ventricular mass index (LVMI) measured by cardiac ultrasonic indices, HRT parameters, including the original heart rate (TO) and slope coeffificient (TS), systolic and diastolic blood pressures (SBP and DBP), as well as blood cholesterol level (TC) were measured before and after treatment. RESULTS After treatment, TO and LVMI were lowered, while TS increased in both the VS group and the CM group (P<0.01), but changed insignificantly in the control group. Significant differences between the CM group and the control group were shown in terms of TO, LVMI, SBP, DBP and TS (P<0.01); and between the CM group and the VS group in terms of TO, LVMI and TS (P<0.01). Moreover, HRT parameters showed an evident correlation with LVMI (r=0.519-0.635, P<0.01). CONCLUSION Combined therapy with XZK and Valsartan can improve hypertensive LVH and HRT parameters, and lessen the damage on the autonomous nervous system.
Collapse
Affiliation(s)
- Chun Gong
- The Cardiovascular Department, the First Affiliated Hospital of Nanchang University, Nanchang (330006), China
| | | | | | | | | | | | | |
Collapse
|
34
|
Bloom HL, Shukrullah I, Veledar E, Gutmann R, London B, Dudley SC. Statins Decrease Oxidative Stress and ICD Therapies. Cardiol Res Pract 2010; 2010:253803. [PMID: 20369058 PMCID: PMC2847377 DOI: 10.4061/2010/253803] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Revised: 01/20/2010] [Accepted: 01/27/2010] [Indexed: 11/20/2022] Open
Abstract
Recent studies demonstrate that statins decrease ventricular arrhythmias in internal cardioverter defibrillator (ICD) patients. The mechanism is unknown, but evidence links increased inflammatory and oxidative states with increased arrhythmias. We hypothesized that statin use decreases oxidation. Methods. 304 subjects with ICDs were surveyed for ventricular arrhythmia. Blood was analyzed for derivatives of reactive oxygen species (DROMs) and interleukin-6 (IL-6). Results. Subjects included 252 (83%) men, 58% on statins, 20% had ventricular arrhythmias. Average age was 63 years and ejection fraction (EF) 20%. ICD implant duration was 29 ± 27 months. Use of statins correlated with lower ICD events (r = 0.12, P = .02). Subjects on statins had lower hsCRP (5.2 versus 6.3; P = .05) and DROM levels (373 versus 397; P = .03). Other factors, including IL-6 and EF did not differ between statin and nonstatin use, nor did beta-blocker or antiarrhythmic use. Multivariate cross-correlation analysis demonstrated that DROMs, statins, IL-6 and EF were strongly associated with ICD events. Multivariate regression shows DROMs to be the dominant predictor. Conclusion. ICD event rate correlates with DROMs, a measure of lipid peroxides. Use of statins is associated with reduced DROMs and fewer ICD events, suggesting that statins exert their effect through reducing oxidation.
Collapse
Affiliation(s)
- Heather L Bloom
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
35
|
Shyu KG, Wang BW, Chen WJ, Kuan P, Hung CR. Mechanism of the inhibitory effect of atorvastatin on endoglin expression induced by transforming growth factor-β1 in cultured cardiac fibroblasts. Eur J Heart Fail 2010; 12:219-26. [DOI: 10.1093/eurjhf/hfq011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Kou-Gi Shyu
- Division of Cardiology; Shin Kong Wu Ho-Su Memorial Hospital; Taipei Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine; Taipei Medical University; Taipei Taiwan
| | - Bao-Wei Wang
- Division of Cardiology; Shin Kong Wu Ho-Su Memorial Hospital; Taipei Taiwan
- School of Medicine; Fu-Jen Catholic University; Taipei County Taiwan
| | - Wei-Jan Chen
- First Cardiovascular Division; Chang Gung Memorial Hospital; Linkou Taipei County Taiwan
| | - Peiliang Kuan
- Division of Cardiology; Shin Kong Wu Ho-Su Memorial Hospital; Taipei Taiwan
| | - Chi-Ren Hung
- Division of Cardiovascular Surgery; Shin Kong Wu Ho-Su Memorial Hospital; 95 Wen-Chang Road Taipei Taiwan
| |
Collapse
|
36
|
Baraka A, Mikhail M, Guemei A, El Ghotny S. Effect of Targeting Mitogen-Activated Protein Kinase on Cardiac Remodeling in Rats. J Cardiovasc Pharmacol Ther 2009; 14:339-46. [DOI: 10.1177/1074248409349620] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background: Increasing evidence suggests that the activation of p38 mitogen-activated protein kinase (p38MAPK) plays a role in cardiac remodeling. Targeting p38MAPK using drugs reported to interfere with its phosphorylation, namely statins and all-trans retinoic acid (atRA), might play a role in ameliorating this remodeling. Methods and Results: Cardiac remodeling was induced in male albino rats by chronic inhibition of nitric oxide (NO) synthesis by N-nitro L-arginine methyl ester (L-NAME). Daily oral administration of L-NAME for 4 weeks resulted in the elevation of mean arterial blood pressure (MABP) together with cardiac remodeling evidenced by an increase in left ventricular-body weight ratio together with an increase in cardiac hydroxyproline concentration and a decrease in left ventricular papillary muscle-developed tension. An elevation in cardiac phosphorylated p38MAPK concentration, tumor necrosis factor alpha concentration and in cardiac caspase 3 activity was also observed. Administration of either rosuvastatin or all-trans retinoic acid (atRA), starting 4 weeks after L-NAME administration, ameliorated remodeling and improved all studied parameters. Conclusions: Targeting MAPK might represent a useful therapeutic avenue to ameliorate cardiac remodeling and support the notion that atRA and statins are potential candidates for the prevention and therapy of cardiac remodeling.
Collapse
Affiliation(s)
- Azza Baraka
- Department of Clinical Pharmacology, Alexandria University, Alexandria, Egypt,
| | - Maher Mikhail
- Department of Clinical Pharmacology, Alexandria University, Alexandria, Egypt
| | - Aida Guemei
- Department of Clinical Pharmacology, Alexandria University, Alexandria, Egypt
| | - Samar El Ghotny
- Department of Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
37
|
Abstract
The number of well-controlled hypertensives is unacceptably low worldwide. Respecting the circadian variation of blood pressure, nontraditional antihypertensives, and treatment in early stages of hypertension are potential ways to improve hypertension therapy. First, prominent variations in circadian rhythm are characteristic for blood pressure. The revolutionary MAPEC (Ambulatory Blood Pressure Monitoring and Cardiovascular Events) study, in 3000 adult hypertensives investigates, whether chronotherapy influences the cardiovascular prognosis beyond blood pressure reduction per se. Second, melatonin, statins and aliskiren are hopeful drugs for hypertension treatment. Melatonin, through its scavenging and antioxidant effects, preservation of NO availability, sympatholytic effect or specific melatonin receptor activation exerts antihypertensive and anti-remodeling effects and may be useful especially in patients with nondipping nighttime blood pressure pattern or with nocturnal hypertension and in hypertensives with left ventricular hypertrophy (LVH). Owing to its multifunctional physiological actions, this indolamine may offer cardiovascular protection far beyond its hemodynamic benefit. Statins exert several pleiotropic effects through inhibition of small guanosine triphosphate-binding proteins such as Ras and Rho. Remarkably, statins reduce blood pressure in hypertensive patients and more importantly they attenuate LVH. Addition of statins should be considered for high-risk hypertensives, for hypertensives with LVH, and possibly for high-risk prehypertensive patients. The direct renin inhibitor, aliskiren, inhibits catalytic activity of renin molecules in circulation and in the kidney, thus lowering angiotensin II levels. Furthermore, aliskiren by modifying the prorenin conformation may prevent prorenin activation. At present, aliskiren should be considered in hypertensive patients not sufficiently controlled or intolerant to other inhibitors of renin-angiotensin system. Third, TROPHY (Trial of Preventing Hypertension) is the first pharmacological intervention for prehypertensive patients revealing that treatment with angiotensin II type 1 receptor blocker attenuates hypertension development and thus decreases the risk of cardiovascular events.
Collapse
Affiliation(s)
- Fedor Simko
- Department of Pathophysiology, Comenius University, Bratislava, Slovak Republic
- 3rd Clinic of Medicine, School of Medicine, Comenius University, Bratislava, Slovak Republic
| | - Olga Pechanova
- Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic
- Institute of Physiology and Center of Cardiovascular Research, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
38
|
Nevzorov R, Novack V, Henkin Y, Kobal SL, Jotkowitz A, Porath A. Discrepancy between results of randomized control studies and retrospective analysis: the case of statin therapy effect on one-year mortality in patients with decompensated heart failure. Eur J Intern Med 2009; 20:494-8. [PMID: 19712852 DOI: 10.1016/j.ejim.2009.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Revised: 12/28/2008] [Accepted: 03/04/2009] [Indexed: 11/27/2022]
Abstract
BACKGROUND In view of the recent reports on a lack of protective effect of statin therapy on clinical outcomes in patients with heart failure, the present study investigated the impact of statin therapy before admission on one-year survival of patients hospitalized due to decompensated heart failure in two groups of patients; with and without ischemic heart disease. METHODS We performed a retrospective cohort analysis of 887 consecutive patients older than 18 years hospitalized with decompensated heart failure between 11/December 2001 and 06/June 2005. Two groups of patients were compared: those who received statins within 3 months before the admission (S) and those who did not (NS). The primary outcome was one-year all-cause mortality. To adjust for a potential misbalance between S and NS groups in baseline characteristics, a propensity score for statin therapy was incorporated into the survival model. RESULTS Two hundred eighty-one patients (31.7%) received statins prior to admission. Patients with ischemic heart disease (IHD) (656/887 subjects, 74%) had higher rate of S therapy as compared to the rest 36.3% vs. 18.6%, p<0.001. Overall one-year mortality rate in the S group was 21% vs. 31.8% in the NS group, p<0.001. In the subgroup of patients with IHD, statins were protective after adjustment for comorbidities and propensity score (hazard ratio [HR], 0.66; 95% CI 0.4-0.97), but in patients with non-ischemic HF statin therapy was not associated with a protective effect (HR 0.77; 95% CI 0.38-1.6). CONCLUSIONS In our study statins' protective effect on one-year survival in HF patients is restricted to patients with IHD. This stands in disagreement with the results of the randomized trials showing no effect of statin therapy. Statin use may be a marker of better health care and despite an extensive adjustment for baseline characteristics, unaccounted bias inherent to retrospective studies may explain the discrepancy in the results.
Collapse
Affiliation(s)
- Roman Nevzorov
- Department of Internal Medicine F, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | | | | | | | | | | |
Collapse
|
39
|
Rosuvastatin protects against angiotensin II-induced renal injury in a dose-dependent fashion. J Hypertens 2009; 27:599-605. [PMID: 19262227 DOI: 10.1097/hjh.0b013e32831ef369] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We showed earlier that statin treatment ameliorates target-organ injury in a transgenic model of angiotensin (Ang) II-induced hypertension. We now test the hypothesis that rosuvastatin (1, 10, and 50 mg/kg/day) influences leukocyte adhesion and infiltration, prevents induction of inducible nitric oxide synthase (iNOS), and ameliorates target-organ damage in a dose-dependent fashion. METHODS We treated rats harboring the human renin and human angiotensinogen genes (dTGR) from week 4 to 8 (n = 20 per group). Untreated dTGR developed severe hypertension, cardiac hypertrophy, and renal damage, with a 100-fold increased albuminuria and focal cortical necrosis. Mortality of untreated dTGR at age 8 weeks was 59%. RESULTS Rosuvastatin treatment decreased mortality dose-dependently. Blood pressure was not affected. Albuminuria was reduced dose-dependently. Interstitial adhesion molecule (ICAM)-1 expression was markedly reduced by rosuvastatin, as were neutrophil and monocyte infiltration. Immunohistochemistry showed an increased endothelial and medial iNOS expression in small vessels, infiltrating cells, afferent arterioles, and glomeruli of dTGR. Immunoreactivity was stronger in cortex than medulla. Rosuvastatin markedly reduced the iNOS expression in both cortex and medulla. Finally, matrix protein (type IV collagen, fibronectin) expression was also dose- dependently reduced by rosuvastatin. CONCLUSION Our findings indicate that rosuvastatin dose- dependently ameliorates angiotensin II-induced-organ damage and almost completely prevents inflammation at the highest dose. The data implicate 3-hydroxy-3-methylglutaryl coenzyme A function in signaling events leading to target-organ damage.
Collapse
|
40
|
Guo WG, Su FF, Yuan LJ, Yang GD, Shi XQ, Li RY, Shu Q, Liu XT, Lu ZF, Zheng QS. Simvastatin inhibits angiotensin II-induced cardiac cell hypertrophy: role of Homer 1a. Clin Exp Pharmacol Physiol 2009; 37:40-5. [PMID: 19515066 DOI: 10.1111/j.1440-1681.2009.05221.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. The scaffolding protein Homer 1a is constitutively expressed in the myocardium, although its function in cardiomyocytes remains poorly understood. The aim of the present study was to investigate Homer 1a expression in hypertrophic cardiac cells and its role in angiotensin (Ang) II-induced cardiac hypertrophy. 2. After serum starvation for 24 h, cells were treated with 1 micromol/L simvastatin, 100 nmol/L angiotensin (Ang) II or their combination added to Dulbecco's modified Eagle's medium containing 0.5% serum. For combination treatment with AngII plus simvastatin, cells were exposed to simvastatin 12 h before the addition of AngII to the medium and cells were then incubated in the presence of both drugs for a further 24 h. Western blotting was used to determine Homer 1a protein expression. Hypertrophy was evaluated by determining the protein content per cell. 3. Homer 1a protein levels were upregulated following AngII-induced hypertrophy in H9C2 cells and neonatal rat cardiomyocytes, and these increases were augmented by simvastatin pretreatment. Concomitantly, simvastatin pretreatment inhibited extracellular signal-regulated kinase (ERK) 1/2 phosphorylation and AngII-induced hypertrophy. 4. The inhibitory effects of simvastatin against AngII-induced hypertrophy were attenuated by Homer 1a silencing, suggesting that simvastatin suppresses cardiac hypertrophy in a Homer 1a-dependent manner. Furthermore, AngII-induced hypertrophy and ERK1/2 phosphorylation in neonatal rat cardiomyocytes were significantly inhibited following the overexpression of Homer 1a using an adenovirus. 5. These results suggest a possible role for Homer 1a in inhibiting cardiac hypertrophy perhaps in part through inhibition of ERK1/2 activation.
Collapse
Affiliation(s)
- Wan-Gang Guo
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zhou Q, Liao JK. Statins and cardiovascular diseases: from cholesterol lowering to pleiotropy. Curr Pharm Des 2009; 15:467-78. [PMID: 19199975 DOI: 10.2174/138161209787315684] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Statins are 3-hydroxy-3-methyglutaryl coenzyme A (HMG-CoA) reductase inhibitors, which are prescribed extensively for cholesterol lowering in the primary and secondary prevention of cardiovascular disease. Recent compelling evidence suggests that the beneficial effects of statins may not only be due to their cholesterol lowering effects, but also, to their cholesterol-independent or pleiotropic effects. Through these so-called pleiotropic effects, statins are directly involved in restoring or improving endothelial function, attenuating vascular remodeling, inhibiting vascular inflammatory response, and perhaps, stabilizing atherosclerotic plaques. These cholesterol-independent effects of statins are predominantly due to their ability to inhibit isoprenoid synthesis, the products of which are important lipid attachments for intracellular signaling molecules, such as Rho, Rac and Cdc42. In particular, inhibition of Rho and its downstream target, Rho-associated coiled-coil containing protein kinase (ROCK), has emerged as the principle mechanisms underlying the pleiotropic effects of statins. This review provides an update of statin-mediated vascular effects beyond cholesterol lowering and highlights recent findings from bench to bedside to support the concept of statin pleiotropy.
Collapse
Affiliation(s)
- Qian Zhou
- Vascular Medicine Research Unit, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02139, USA
| | | |
Collapse
|
42
|
Naito M, Shenoy A, Aoyama I, Koopmeiners JS, Komers R, Schnaper HW, Bomsztyk K. High ambient glucose augments angiotensin II-induced proinflammatory gene mRNA expression in human mesangial cells: effects of valsartan and simvastatin. Am J Nephrol 2009; 30:99-111. [PMID: 19225232 DOI: 10.1159/000203619] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Accepted: 01/16/2009] [Indexed: 12/28/2022]
Abstract
BACKGROUND Hyperglycemia may potentiate the adverse renal effects of angiotensin II (AII). In the kidney, the major target of AII action is the glomerular mesangial cell, where its hemodynamic and proinflammatory action contributes to renal injury. AII action is mediated by several types of cell receptors. Among those, the AT1 receptor has been best studied using specific AII receptor blockers (ARBs). These agents have emerged as major new modalities in the prevention and amelioration of renal disease where the ARB renoprotective anti-inflammatory properties could be more important than previously appreciated. Like the ARBs, statins may also modulate inflammatory responses that are renoprotective and complement their cholesterol-lowering effects. AIM The aim of this project was to (i) identify a repertoire of proinflammatory mesangial cell AII-inducible mRNAs; (ii) determine if the AII-induced proinflammatory mRNA responses depend on ambient glucose, and (iii) test the anti-inflammatory effectiveness of an ARB, valsartan, either alone or in combination with a statin, simvastatin. RESULTS/CONCLUSIONS Using high-density microarrays and real-time PCR we identified several AII-inducible proinflammatory mesangial genes that exhibited augmented mRNA responses in high-glucose milieu. Valsartan blocked the AII-induced mRNA expression of proinflammatory genes (i.e. MCP-1, LIF and COX-2) maintained in normal and high glucose. These observations add to the mounting evidence that ARBs have anti-inflammatory effects in the kidney, a beneficial effect that may be more important in protecting renal function in diabetic patients. While simvastatin inhibited expression of some mRNAs encoding chemokines/cytokines, it enhanced expression of mRNA encoding COX-2, a key mediator of inflammation. Thus, the non-cholesterol effects of statins on inflammatory responses appear complex.
Collapse
Affiliation(s)
- Masayo Naito
- UW Medicine Lake Union Research, University of Washington, Seattle, WA 98109, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Kishi T, Yamada A, Okamatsu S, Sunagawa K. Atorvastatin might improve ventricular electrostability and decelerate the deterioration of renal function in patients with heart failure and diabetes mellitus. J Cardiol 2009; 53:341-8. [PMID: 19477374 DOI: 10.1016/j.jjcc.2008.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 11/09/2008] [Accepted: 12/03/2008] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND PURPOSE Previous studies suggested that statins have pleiotropic effects, such as improvements in endothelial function, as well as anti-inflammatory, anti-proliferative, and anti-oxidative effects. These effects might benefit patients with heart failure. In those patients, statins relieved symptoms, decreased the frequency of hospitalization, suppressed neurohumoral activation, and improved cardiac function. However, it remains unknown how statins impact pathophysiology of heart failure with diabetes mellitus. The aim of this study was to investigate the effects of atorvastatin on pathophysiology of heart failure with diabetes mellitus. METHODS AND RESULTS We enrolled retrospectively 128 patients with heart failure with diabetes mellitus who were admitted from January 2003 to December 2005. Among these patients, 80 received atorvastatin (statin group) and the remaining patients served as controls (non-statin group). At study entry, there were no significant differences in the patient profiles between the two groups except for the low-density lipoprotein cholesterol level being higher in the statin group. After the follow-up period of two years, the frequency of re-hospitalization, brain natriuretic peptide, premature ventricular contractions, Lown grade, and deterioration of glomerular filtration rate were significantly less in the statin group. CONCLUSION Atorvastatin might benefit patients with heart failure and diabetes mellitus by improving ventricular electrical stability and decelerating deterioration of renal function.
Collapse
Affiliation(s)
- Takuya Kishi
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Higashi-ku, Fukuoka, Japan.
| | | | | | | |
Collapse
|
44
|
Cui W, Matsuno K, Iwata K, Ibi M, Katsuyama M, Kakehi T, Sasaki M, Ikami K, Zhu K, Yabe-Nishimura C. NADPH Oxidase Isoforms and Anti-hypertensive Effects of Atorvastatin Demonstrated in Two Animal Models. J Pharmacol Sci 2009; 111:260-8. [DOI: 10.1254/jphs.09148fp] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
45
|
Landmesser U, Wollert KC, Drexler H. Potential novel pharmacological therapies for myocardial remodelling. Cardiovasc Res 2008; 81:519-27. [PMID: 19019834 DOI: 10.1093/cvr/cvn317] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Left ventricular (LV) remodelling remains an important treatment target in patients after myocardial infarction (MI) and chronic heart failure (CHF). Accumulating evidence has supported the concept that beneficial effects of current pharmacological treatment strategies to improve the prognosis in these patients, such as angiotensin-converting enzyme (ACE) inhibition, angiotensin type 1 receptor blocker therapy, and beta-blocker therapy, are related, at least in part, to their effects on LV remodelling and dysfunction. However, despite modern reperfusion therapy after MI and optimized treatment of patients with CHF, LV remodelling is observed in a substantial proportion of patients and is associated with an adverse clinical outcome. These observations call for novel therapeutic strategies to prevent or even reverse cardiac remodelling. Recent insights from experimental studies have provided new targets for interventions to prevent or reverse LV remodelling, i.e. reduced endothelial nitric oxide (NO) synthase-derived NO availability, activation of cardiac and leukocyte-dependent oxidant stress pathways, inflammatory pathway activation, matrix-metalloproteinase activation, or stem cell transfer and delivery of novel paracrine factors. An important challenge in translating these observations from preclinical studies into clinical treatment strategies relates to the fact that clinical studies are designed on top of established pharmacological therapy, whereas most experimental studies have tested novel interventions without concomitant drug regimens such as ACE inhibitors or beta-blockers. Therefore, animal studies may overestimate the effect of potential novel treatment strategies on LV remodelling and dysfunction, since established pharmacological therapies may act, in part, via identical or similar signalling pathways. Nevertheless, preclinical studies provide essential information for identifying potential novel targets, and their potential drawbacks, and are required for developing novel clinical treatment strategies to prevent or reverse LV remodelling and dysfunction.
Collapse
Affiliation(s)
- Ulf Landmesser
- Deparment of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany
| | | | | |
Collapse
|
46
|
Osorio JC, Cheema FH, Martens TP, Mahmut N, Kinnear C, Gonzalez AMD, Bonney W, Homma S, Liao JK, Mital S. Simvastatin reverses cardiac hypertrophy caused by disruption of the bradykinin 2 receptor. Can J Physiol Pharmacol 2008; 86:633-42. [PMID: 18758513 DOI: 10.1139/y08-068] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Bradykinin 2 receptor (B2R) deficiency predisposes to cardiac hypertrophy and hypertension. The pathways mediating these effects are not known. Two-month-old B2R knockout (KO) and wild-type (WT) mice were assigned to 4 treatment groups (n = 12-14/group): control (vehicle); nitro-L-arginine methyl ester (L-NAME) an NO synthase inhibitor; simvastatin (SIM), an NO synthase activator; and SIM+L-NAME. Serial echocardiography was performed and blood pressure (BP) at 6 weeks was recorded using a micromanometer. Myocardial eNOS and mitogen-activated protein kinase (MAPK, including ERK, p38, and JNK) protein expression were measured. Results showed that (i) B2RKO mice had significantly lower ejection fraction than did WT mice (61% +/- 1% vs. 73% +/- 1%), lower myocardial eNOS and phospho-eNOS, normal systolic BP, and higher LV mass, phospho-p38, and JNK; (ii) L-NAME increased systolic BP in KO mice (117 +/- 19 mm Hg) but not in WT mice and exacerbated LV hypertrophy and dysfunction; and (iii) in KO mice, SIM decreased hypertrophy, p38, and JNK, improved function, increased capillary eNOS and phospho-eNOS, and prevented L-NAME-induced LV hypertrophy without lowering BP. We conclude that disruption of the B2R causes maladaptive cardiac hypertrophy with myocardial eNOS downregulation and MAPK upregulation. SIM reverses these abnormalities and prevents the development of primary cardiac hypertrophy as well as hypertrophy secondary to L-NAME-induced hypertension.
Collapse
|
47
|
|
48
|
Abstract
Although hypertensive heart disease (HHD) is clinically characterized 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 (eg, fibrosis). Myocardial fibrosis is the major determinant of diastolic dysfunction/failure in patients with HHD. Recent available data on the determination of serum concentrations of collagen-derived serum peptides, as well as 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 using specific pharmacological agents in patients with HHD.
Collapse
Affiliation(s)
- Javier Díez
- Division of Cardiovascular Sciences, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain.
| |
Collapse
|
49
|
Abstract
The syndrome of heart failure is characterized by increased levels of circulating inflammatory mediators, which have been implicated in the pathogenesis of heart failure. Recently, a number of studies have suggested that statins may exert salutary effects in patients who have heart failure by virtue of their pleiotropic (non-lipid lowering) actions. This article focuses on the non-lipid lowering effects of statins, with an emphasis on the anti-inflammatory properties of these agents.
Collapse
|
50
|
Abstract
Sympathetic nervous system activation in heart failure, as indexed by elevated norepinephrine levels, higher muscle sympathetic nerve activity and reduced heart rate variability, is associated with pathologic ventricular remodeling, increased arrhythmias, sudden death, and increased mortality. Recent evidence suggests that 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor (statin) therapy may provide survival benefit in heart failure of both ischemic and nonischemic etiology, and one potential mechanism of benefit of statins in heart failure is modulation of the autonomic nervous system. Animal models of heart failure demonstrate reduced sympathetic activation and improved sympathovagal balance with statin therapy. Initial human studies have reported mixed results. Ongoing translational studies and outcomes trials will help delineate the potentially beneficial effects of statins on the autonomic nervous system in heart failure.
Collapse
|