1
|
Mamazhakypov A, Sartmyrzaeva M, Sarybaev AS, Schermuly R, Sydykov A. Clinical and Molecular Implications of Osteopontin in Heart Failure. Curr Issues Mol Biol 2022; 44:3573-3597. [PMID: 36005141 PMCID: PMC9406846 DOI: 10.3390/cimb44080245] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
The matricellular protein osteopontin modulates cell-matrix interactions during tissue injury and healing. A complex multidomain structure of osteopontin enables it not only to bind diverse cell receptors but also to interact with various partners, including other extracellular matrix proteins, cytokines, and growth factors. Numerous studies have implicated osteopontin in the development and progression of myocardial remodeling in diverse cardiac diseases. Osteopontin influences myocardial remodeling by regulating extracellular matrix production, the activity of matrix metalloproteinases and various growth factors, inflammatory cell recruitment, myofibroblast differentiation, cardiomyocyte apoptosis, and myocardial vascularization. The exploitation of osteopontin loss- and gain-of-function approaches in rodent models provided an opportunity for assessment of the cell- and disease-specific contribution of osteopontin to myocardial remodeling. In this review, we summarize the recent knowledge on osteopontin regulation and its impact on various cardiac diseases, as well as delineate complex disease- and cell-specific roles of osteopontin in cardiac pathologies. We also discuss the current progress of therapeutics targeting osteopontin that may facilitate the development of a novel strategy for heart failure treatment.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Department of Internal Medicine, German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Meerim Sartmyrzaeva
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek 720040, Kyrgyzstan
| | - Akpay Sh. Sarybaev
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek 720040, Kyrgyzstan
| | - Ralph Schermuly
- Department of Internal Medicine, German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Akylbek Sydykov
- Department of Internal Medicine, German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
- Correspondence:
| |
Collapse
|
2
|
Osteopontin in Cardiovascular Diseases. Biomolecules 2021; 11:biom11071047. [PMID: 34356671 PMCID: PMC8301767 DOI: 10.3390/biom11071047] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Unprecedented advances in secondary prevention have greatly improved the prognosis of cardiovascular diseases (CVDs); however, CVDs remain a leading cause of death globally. These findings suggest the need to reconsider cardiovascular risk and optimal medical therapy. Numerous studies have shown that inflammation, pro-thrombotic factors, and gene mutations are focused not only on cardiovascular residual risk but also as the next therapeutic target for CVDs. Furthermore, recent clinical trials, such as the Canakinumab Anti-inflammatory Thrombosis Outcomes Study trial, showed the possibility of anti-inflammatory therapy for patients with CVDs. Osteopontin (OPN) is a matricellular protein that mediates diverse biological functions and is involved in a number of pathological states in CVDs. OPN has a two-faced phenotype that is dependent on the pathological state. Acute increases in OPN have protective roles, including wound healing, neovascularization, and amelioration of vascular calcification. By contrast, chronic increases in OPN predict poor prognosis of a major adverse cardiovascular event independent of conventional cardiovascular risk factors. Thus, OPN can be a therapeutic target for CVDs but is not clinically available. In this review, we discuss the role of OPN in the development of CVDs and its potential as a therapeutic target.
Collapse
|
3
|
Yasui H, Takahama H, Kanzaki H, Ishibashi-Ueda H, Morita N, Sugano Y, Hasegawa T, Ohara T, Amaki M, Funada A, Yoshikawa Y, Yasuda S, Ogawa H, Anzai T. Time-Course Changes of Cardiac-Specific Inflammation in a Patient With Left Ventricular Calcified Amorphous Tumor. Circ J 2015; 79:2069-71. [PMID: 26062949 DOI: 10.1253/circj.cj-15-0136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Hiroki Yasui
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Dalal S, Zha Q, Daniels CR, Steagall RJ, Joyner WL, Gadeau AP, Singh M, Singh K. Osteopontin stimulates apoptosis in adult cardiac myocytes via the involvement of CD44 receptors, mitochondrial death pathway, and endoplasmic reticulum stress. Am J Physiol Heart Circ Physiol 2014; 306:H1182-91. [PMID: 24531809 DOI: 10.1152/ajpheart.00954.2013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Increased osteopontin (OPN) expression associates with increased myocyte apoptosis and myocardial dysfunction. The objective of this study was to identify the receptor for OPN and get insight into the mechanism by which OPN induces cardiac myocyte apoptosis. Adult rat ventricular myocytes (ARVMs) and transgenic mice expressing OPN in a myocyte-specific manner were used for in vitro and in vivo studies. Treatment with purified OPN (20 nM) protein or adenoviral-mediated OPN expression induced apoptosis in ARVMs. OPN co-immunoprecipitated with CD44 receptors, not with β1 or β3 integrins. Proximity ligation assay confirmed interaction of OPN with CD44 receptors. Neutralizing anti-CD44 antibodies inhibited OPN-stimulated apoptosis. OPN activated JNKs and increased expression of Bax and levels of cytosolic cytochrome c, suggesting involvement of mitochondrial death pathway. OPN increased endoplasmic reticulum (ER) stress, as evidenced by increased expression of Gadd153 and activation of caspase-12. Inhibition of JNKs using SP600125 or ER stress using salubrinal or caspase-12 inhibitor significantly reduced OPN-stimulated apoptosis. Expression of OPN in adult mouse heart in myocyte-specific manner associated with decreased left ventricular function and increased myocyte apoptosis. In the heart, OPN expression increased JNKs and caspase-12 activities, and expression of Bax and Gadd153. Thus, OPN, acting via CD44 receptors, induces apoptosis in myocytes via the involvement of mitochondrial death pathway and ER stress.
Collapse
Affiliation(s)
- Suman Dalal
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Umar S, Nadadur R, Iorga A, Amjedi M, Matori H, Eghbali M. Cardiac structural and hemodynamic changes associated with physiological heart hypertrophy of pregnancy are reversed postpartum. J Appl Physiol (1985) 2012; 113:1253-9. [PMID: 22923507 DOI: 10.1152/japplphysiol.00549.2012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Pregnancy is associated with ventricular hypertrophy and volume overload. Here we investigated whether late pregnancy is associated with cardiac structural and hemodynamic changes, and if these changes are reversed postpartum. Female mice (C57BL/6) were used in nonpregnant diestrus (NP), late-pregnant (LP), or 7-day postpartum (PP7) stages. Echocardiography and cardiac catheterization were performed to monitor cardiac hemodynamics. Transcript expression of proangiogenic vascular endothelial growth factor, cardiac fetal gene osteopontin, cardiac extracellular matrix-degrading enzymes matrix metalloproteinase-2, and a disintegrin and metalloproteinase-15 and -17 were assessed by RT-PCR. Masson trichrome staining for cardiac fibrosis and endothelial marker CD31 immunostaining for angiogenesis were performed. Heart hypertrophy in LP was fully reversed in PP7 (heart weight: NP = 114 ± 4 mg; LP = 147 ± 2 mg; PP7 = 117 ± 8 mg, P < 0.05 for LP vs. PP7). LP had elevated left ventricular (LV) pressure (119 ± 5 mmHg in LP vs. 92 ± 7 mmHg in NP, P < 0.05) that was restored at PP7 (95 ± 8 mmHg, P < 0.001 vs. LP). LP had increased LV contractility (maximal rate of increase of LV pressure = 6,664 ± 297 mmHg/s in LP vs. 4,294 ± 568 mmHg/s in NP, P < 0.01) that was restored at PP7 (5,313 ± 636 mmHg/s, P < 0.05 vs. LP). LV ejection fraction was reduced in LP (LP = 58 ± 1% vs. NP = 70 ± 4%, P < 0.001) and was already restored at PP1 (77 ± 2%, P < 0.001 vs. LP). Myocardial angiogenesis was significantly increased in LP (capillary density = 1.25 ± 0.02 vs. 0.95 ± 0.01 capillaries/myocyte in NP, P < 0.001) and was fully restored in PP7 (0.98 ± 0.01, P < 0.001 vs. LP). Vascular endothelial growth factor was upregulated in LP (LP = 1.4 ± 0.1 vs. NP = 1 ± 0.1, normalized to NP, P < 0.001) and was restored in PP7 (PP7 = 0.83 ± 0.1, P < 0.001 vs. LP). There was no increase in cardiac fibrosis in LP. Matrix metalloproteinase-2 transcript levels were downregulated in LP (LP = 0.47 ± 0.03 vs. NP = 1 ± 0.01, normalized to NP, P < 0.001) and was restored at PP7 (0.70 ± 0.1, P < 0.001 vs. LP). In conclusion, pregnancy-induced heart hypertrophy is associated with transient cardiac dysfunction, increased cardiac angiogenesis, lack of fibrosis, and decreased expression of remodeling enzymes that are reversed postpartum.
Collapse
Affiliation(s)
- Soban Umar
- Department of Anesthesiology, Division of Molecular Medicine David Geffen School of Medicine at University of California-Los Angeles, CA 90095, USA
| | | | | | | | | | | |
Collapse
|
6
|
Nadadur RD, Umar S, Wong G, Eghbali M, Iorga A, Matori H, Partow-Navid R, Eghbali M. Reverse right ventricular structural and extracellular matrix remodeling by estrogen in severe pulmonary hypertension. J Appl Physiol (1985) 2012; 113:149-58. [PMID: 22628376 DOI: 10.1152/japplphysiol.01349.2011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Chronic pulmonary hypertension (PH) leads to right-ventricular failure (RVF) characterized by RV remodeling. Ventricular remodeling is emerging as an important process during heart failure and recovery. Remodeling in RVF induced by PH is not fully understood. Recently we discovered that estrogen (E2) therapy can rescue severe preexisting PH. Here, we focused on whether E2 (42.5 μg·kg(-1)·day(-1), 10 days) can reverse adverse RV structural and extracellular matrix (ECM) remodeling induced by PH using monocrotaline (MCT, 60 mg/kg). RV fibrosis was evident in RVF males. Intact females developed less severe RV remodeling compared with males and ovariectomized (OVX) females. Novel ECM-degrading disintegrin-metalloproteinases ADAM15 and ADAM17 transcripts were elevated ∼2-fold in all RVF animals. E2 therapy reversed RV remodeling in all groups. In vitro, E2 directly inhibited ANG II-induced expression of fibrosis markers as well as the metalloproteinases in cultured cardiac fibroblasts. Estrogen receptor-β agonist diarylpropionitrile (DPN) but not estrogen receptor-α agonist 4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT) was as effective as E2 in inhibiting expression of these genes. Expression of ECM-interacting cardiac fetal-gene osteopontin (OPN) also increased ∼9-fold in RVF males. Intact females were partially protected from OPN upregulation (∼2-fold) but OVX females were not. E2 reversed OPN upregulation in all groups. Upregulation of OPN was also reversed in vitro by E2. Plasma OPN was elevated in RVF (∼1.5-fold) and decreased to control levels in the E2 group. RVF resulted in elevated Akt phosphorylation, but not ERK, in the RV, and E2 therapy restored Akt phosphorylation. In conclusion, E2 therapy reverses adverse RV remodeling associated with PH by reversing fibrosis and upregulation of novel ECM enzymes ADAM15, ADAM17, and OPN. These effects are likely mediated through estrogen receptor-β.
Collapse
Affiliation(s)
- Rangarajan D Nadadur
- Department of Anesthesiology, Division of Molecular Medicine, University of California at Los Angeles, Los Angeles, California 90095-7115, USA
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Niebroj-Dobosz I, Madej-Pilarczyk A, Marchel M, Sokołowska B, Hausmanowa-Petrusewicz I. Osteopontin – a fibrosis-related marker – in dilated cardiomyopathy in patients with Emery-Dreifuss muscular dystrophy. Scandinavian Journal of Clinical and Laboratory Investigation 2011; 71:658-62. [DOI: 10.3109/00365513.2011.619272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
8
|
Okamoto H, Imanaka-Yoshida K. Matricellular proteins: new molecular targets to prevent heart failure. Cardiovasc Ther 2011; 30:e198-209. [PMID: 21884011 DOI: 10.1111/j.1755-5922.2011.00276.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Matricellular proteins are highly expressed in reparative responses to pressure and volume overload, ischemia, oxidative stress after myocardial injury, and modulate the inflammatory and fibrotic process in ventricular remodeling, which leads to cardiac dysfunction and eventually overt heart failure. Generally, matricellular proteins loosen strong adhesion of cardiomyocytes to extracellular matrix, which would help cells to move for rearrangement and allow inflammatory cells and capillary vessels to spread during tissue remodeling. Among matricellular proteins, osteopontin (OPN) and tenascin-C (TN-C) are de-adhesion proteins and upregulate the expression and activity of matrix metalloproteinases. These matricellular proteins could be key molecules to diagnose cardiac remodeling and also might be targets for the prevention of adverse ventricular remodeling. This review provides an overview of the role of matricellular proteins such as OPN and TN-C in cardiac function and remodeling, as determined by both in basic and in clinical studies.
Collapse
Affiliation(s)
- Hiroshi Okamoto
- Department of Cardiovascular Medicine, Hokkaido Medical Center, Sapporo, Japan. okamotoh@ med.hokudai.ac.jp
| | | |
Collapse
|
9
|
Francia P, Balla C, Ricotta A, Uccellini A, Frattari A, Modestino A, Borro M, Simmaco M, Salvati A, De Biase L, Volpe M. Plasma osteopontin reveals left ventricular reverse remodelling following cardiac resynchronization therapy in heart failure. Int J Cardiol 2010; 153:306-10. [PMID: 20863582 DOI: 10.1016/j.ijcard.2010.08.048] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2010] [Revised: 06/18/2010] [Accepted: 08/17/2010] [Indexed: 11/17/2022]
Abstract
BACKGROUND Cardiac resynchronization therapy (CRT) promotes left ventricular (LV) reverse remodelling and affects myocardial collagen turnover in heart failure (HF) patients. Osteopontin (OPN) is a matrix glycoprotein required for the activation of fibroblasts upon TGF-β1 stimulation. In humans, plasma OPN and OPN-expressing lymphocytes correlate with the severity of HF. We sought to evaluate whether plasma OPN and TGF-β1 reflect LV reverse remodelling following CRT. METHODS Eighteen patients (12 men, mean age 65 ± 11 years) undergoing CRT were studied. Patients underwent baseline clinical and echocardiographic evaluation, and assessment of plasma OPN and TGF-β1. The evaluation was repeated 8.5 ± 4 months after device implantation. Eight healthy age- and sex-matched subjects served as controls. RESULTS In HF patients, baseline plasma OPN and TGF-β1 were higher as compared to control subjects (OPN: 99 ± 48 vs 59 ± 22 ng/ml; p<0.05; TGF-β1: 15.9 ± 8.0 vs 9.3 ± 5.6 ng/ml; p<0.05). At follow-up, 12 patients responded to CRT and showed LV reverse remodelling, whereas 6 did not. Plasma OPN decreased in CRT responders (108 ± 47 vs 84 ± 37 ng/ml; p=0.03) and increased in non-responders (79 ± 58 vs 115 ± 63 ng/ml; p<0.01). TGF-β1 showed a trend towards reduction in responders (17.5 ± 8.7 vs 10.2 ± 8.9 ng/ml; p=0.08) and was unchanged in non-responders. A significant correlation (r=-0.56; p=0.01) was found between relative changes of LVESV and plasma OPN. CONCLUSIONS CRT-induced LV reverse remodelling is reflected by changes in plasma OPN. Circulating OPN may represent a marker of LV dilation/impairment and an indicator of the response to HF therapies promoting LV reverse remodelling.
Collapse
Affiliation(s)
- Pietro Francia
- Cardiology, 2nd Faculty of Medicine, Sapienza University, Sant'Andrea Hospital, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
Osteopontin (OPN), also known as 44kDa bone phosphoprotein, sialoprotein I, secreted phosphoprotein I, 2ar, uropontin, and early T-lymphocyte activation-1 (Eta-1), is a multifunctional protein. OPN has been found to be expressed in various cell types and species with many physiologic and pathologic functions. OPN has emerged as a potential biomarker and mediator in cardiovascular disease. In this review, we will discuss the roles of OPN in cardiovascular disease, specifically in vascular and valvular heart disease, myocardial infarction and heart failure.
Collapse
|
11
|
|
12
|
Increased plasma levels of osteopontin are associated with activation of the renin-aldosterone system and with myocardial and coronary microvascular damage in dilated cardiomyopathy. Cytokine 2009; 49:325-30. [PMID: 20031444 DOI: 10.1016/j.cyto.2009.11.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 10/28/2009] [Accepted: 11/22/2009] [Indexed: 11/22/2022]
Abstract
In patients with dilated cardiomyopathy (DCM) abnormal myocardial blood flow (MBF) has been associated with coronary microvascular dysfunction. The aim of this study was to test the hypothesis that osteopontin (OPN) plasma levels could be associated with the activation of the renin-aldosterone system (RAS) in these patients and be involved in mediating myocardial and coronary damage. In 66 patients with idiopathic left ventricular dysfunction of variable severity the plasma levels of OPN were correlated with biomarkers of systemic metabolism, RAS activation, myocardial dysfunction and with clinical indexes of left ventricle (LV) function and perfusion obtained by 2D-echocardiography and PET. As compared to controls, patients showed a significant increase of inflammatory markers (OPN: 508+/-30.8ng/ml vs. 426.9+/-16.4, p<0.05 and interleukin (IL)-6: 1.71+/-0.29pg/ml vs. 0.38+/-0.03pg/ml, p<0.001) and of indexes of cardiac damage. OPN levels were significantly correlated with the extent of microvascular dysfunction (MBF at rest: p=0.01; during dipyridamole: p=0.0003) and with plasma renin activity (PRA) (r=0.26, p=0.04). Both in patients with milder or more severe LV dysfunction lower MBF values were associated with higher OPN levels and PRA. These results suggest a interdependent role of RAS and vascular inflammation in cardiomyopathy.
Collapse
|
13
|
Francia P, Uccellini A, Frattari A, Modestino A, Ricotta A, Balla C, Scialla L, Volpe M. Extracellular Matrix Remodelling in Myocardial Hypertrophy and Failure. High Blood Press Cardiovasc Prev 2009; 16:195-9. [DOI: 10.2165/11530450-000000000-00000] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Accepted: 06/26/2009] [Indexed: 11/02/2022] Open
|
14
|
Osteopontin: role in extracellular matrix deposition and myocardial remodeling post-MI. J Mol Cell Cardiol 2009; 48:538-43. [PMID: 19573532 DOI: 10.1016/j.yjmcc.2009.06.015] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 06/08/2009] [Accepted: 06/18/2009] [Indexed: 02/07/2023]
Abstract
Remodeling after myocardial infarction (MI) associates with left ventricular (LV) dilation, decreased cardiac function and increased mortality. The dynamic synthesis and breakdown of extracellular matrix (ECM) proteins play a significant role in myocardial remodeling post-MI. Expression of osteopontin (OPN) increases in the heart post-MI. Evidence has been provided that lack of OPN induces LV dilation which associates with decreased collagen synthesis and deposition. Inhibition of matrix metalloproteinases, key players in ECM remodeling process post-MI, increased ECM deposition (fibrosis) and improved LV function in mice lacking OPN after MI. This review summarizes--1) signaling pathways leading to increased expression of OPN in the heart; 2) the alterations in the structure and function of the heart post-MI in mice lacking OPN; and 3) mechanisms involved in OPN-mediated ECM remodeling post-MI.
Collapse
|
15
|
Soejima H, Irie A, Fukunaga T, Oe Y, Kojima S, Kaikita K, Kawano H, Sugiyama S, Yoshimura M, Kishikawa H, Nishimura Y, Ogawa H. Osteopontin expression of circulating T cells and plasma osteopontin levels are increased in relation to severity of heart failure. Circ J 2008; 71:1879-84. [PMID: 18037740 DOI: 10.1253/circj.71.1879] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND T cells in peripheral blood reflect the systemic inflammatory response in patients with heart failure (HF). In a rat model of HF, osteopontin is dramatically increased in the left ventricular myocardium, so the association between osteopontin and HF was examined in the present study. METHODS AND RESULTS Peripheral blood was collected from 93 patients with heart disease and 38 controls. Left ventricular ejection fraction (LVEF) was calculated using a modified Simpson's rule. The 93 patients were classified into 3 classes according to the New York Heart Association (NYHA) functional classification. Osteopontin-expressing CD4+ T cells were quantified by flow cytometry. Plasma osteopontin levels (ng/ml) and the frequencies of osteopontin-expressing CD4+ T cells (%) were higher in patients with HF than in controls (800+/-554, 575+/-229, p=0.016 and 27.3+/-12.2, 16.7+/-10.0, p<0.001). Furthermore, the plasma osteopontin levels and the frequencies of osteopontin-expressing CD4+ T cells increased in proportion to the severity of the NYHA functional class. The frequencies of osteopontin-expressing CD4+ T cells were significantly correlated with LVEF (r=-0.336, p=0.0048) and log plasma brain natriuretic peptide levels (r=0.305, p=0.0025). CONCLUSIONS Osteopontin expression of circulating CD4+ T cells and plasma osteopontin levels reflect the severity of HF. Osteopontin could be a new target in the assessment of HF.
Collapse
Affiliation(s)
- Hirofumi Soejima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
A matricellular protein, osteopontin (OPN), is expressed in response to mechanical stress and similar stimuli in the heart, integrates the inter-ECM signal transduction network of component cells, and maintains efficient contractility through quantitative and qualitative control of extracellular matrix (ECM) proteins. In particular, OPN is re-expressed in the process of tissue damage; combines with other cell growth factors, cytokines, chemokines, and proteases as a cytokine itself or as an adhesion molecule; and controls the differentiation and growth of cells involved in restoration of tissues by controlling inter-cellular signal transduction and production of ECM proteins through regulation of expression levels and activity. A study using mice lacking a functional OPN gene indicated that tissue restoration fails and collagen deposition is inhibited through matrix metalloproteinases (MMPs) in mice lacking OPN. Thus, while OPN accelerates the cardiovascular remodeling process, it also regulates the balance of various inter-cellular activities. In addition, OPN not only promotes arteriosclerosis but is also closely associated with angiogenesis. With the roles of OPN expected to be clinically elucidated, the clinical use of OPN for control of cardiovascular remodeling may be feasible. Points (1) Osteopontin (OPN) efficiently propagates contraction in the heart as a matricellular protein and thereby controls ECM proteins both quantitatively and qualitatively. (2) The quantitative and qualitative control of ECM proteins is involved in interaction with OPN receptors including those of the integrin family, CD44, and others. (3) OPN promotes myocardial remodeling through TGFbeta and MMPs. (4) OPN not only promotes arteriosclerosis but is also closely associated with arteriosteogenesis. (5) In animals lacking OPN, tissue remodeling process is inhibited, especially in terms of fibrosis after myocardial infarction. (6) While the significance of OPN as an immune system molecule is still unclear in detail, the significance of OPN in the regenerative immune system has begun to be determined.
Collapse
Affiliation(s)
- Hiroshi Okamoto
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Kita-Ku, Kita-14, Nishi-5, Sapporo 060-8638, Japan.
| |
Collapse
|
17
|
Abstract
Myocardial infarction triggers an inflammatory cascade that results in healing and replacement of the damaged tissue with scar. Cardiomyocyte necrosis triggers innate immune mechanisms eliciting Toll-like receptor- mediated responses, activating the complement cascade and generating reactive oxygen species. Subsequent activation of NF-kappaB is a critical element in the regulation of cytokine, chemokine, and adhesion molecule expression in the ischemic myocardium. Chemokine induction mediates leukocyte recruitment in the myocardium. Pleiotropic proinflammatory cytokines, such as TNF-alpha, IL-1, and IL-6, are also upregulated in the infarct and exert a wide range of effects on a variety of cell types. Timely repression of proinflammatory gene synthesis is crucial for optimal healing; IL-10 and TGF-beta-mediated pathways may be important for suppression of chemokine and cytokine expression and for resolution of the leukocytic infiltrate. In addition, TGF-beta may be critically involved in inducing myofibroblast differentiation and activation, promoting extracellular matrix protein deposition in the infarcted area. The composition of the extracellular matrix plays an important role in regulating cell behavior. Both structural and matricellular proteins modulate cell signaling through interactions with specific surface receptors. The molecular and cellular changes associated with infarct healing directly influence ventricular remodeling and affect prognosis in patients with myocardial infarction.
Collapse
|
18
|
Kamiyoshi Y, Takahashi M, Yokoseki O, Yazaki Y, Hirose SI, Morimoto H, Watanabe N, Kinoshita O, Hongo M, Ikeda U. Mycophenolate mofetil prevents the development of experimental autoimmune myocarditis. J Mol Cell Cardiol 2005; 39:467-77. [PMID: 15978615 DOI: 10.1016/j.yjmcc.2005.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2004] [Revised: 03/25/2005] [Accepted: 04/20/2005] [Indexed: 11/19/2022]
Abstract
Experimental autoimmune myocarditis (EAM) is characterized by the appearance of multinucleated giant cells. EAM leads to severe myocardial damage and is a useful model of human giant cell myocarditis. We investigated whether mycophenolate mofetil (MMF), which is a potent immunosuppressant, prevents the development of myocarditis in a rat EAM model, and focused on the role of osteopontin (OPN) in the pathogenesis of this disorder. Adult Lewis rats were immunized with porcine cardiac myosin to establish EAM. The early MMF treatment completely prevented the development of EAM, and the late MMF treatment was also effective even against established EAM. Echocardiogram demonstrated that left ventricular function was also improved by the treatment with MMF. Real-time RT-PCR analysis showed that both early and late MMF treatments significantly inhibited myocarditis-induced OPN mRNA expression in the heart. Immunohistochemistry revealed that OPN expression was prominent in the myocardium on day 14, whereas expression was observed in the infiltrated macrophages on day 21. Mycophenolic acid (MPA) did inhibit agonist-induced OPN expression in cultured cardiomyocytes. These results show the therapeutic potential of MMF for autoimmune myocarditis and provide new insights into the pathogenesis of this disease.
Collapse
Affiliation(s)
- Yuichi Kamiyoshi
- Division of Cardiovascular Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Decompensated heart failure (HF) may be defined as sustained deterioration of at least one New York Heart Association functional class, usually with evidence of sodium retention. Episodes of decompensation are most commonly precipitated by sodium retention, often associated with medication noncompliance. Our therapeutic approach to hospitalized patients is based on the documented hemodynamic responses to vasodilator therapy, with redistribution of mitral regurgitant flow to forward cardiac output and decompression of the left atrium. Invasive hemodynamic monitoring is seldom required for the effective management of patients with HF and there are risks associated with pulmonary artery catheterization. The currently available parenteral vasoactive drugs for decompensated heart failure include: (i) vasodilators such as nesiritide, nitroprusside and nitroglycerin (glyceryl trinitrate); (ii) catecholamine inotropes, primarily dobutamine; and (iii) inodilators such as milrinone, a phosphodiesterase inhibitor. Vasodilators are most appropriate for those patients who are primarily volume-overloaded, but with adequate peripheral perfusion. In this class of agents, nesiritide (recombinant human B-type natriuretic peptide) offers advantages over currently available drugs. Nesiritide produces rapid and sustained decreases in right atrial and pulmonary capillary wedge pressures, with reduction in pulmonary and systemic vascular resistance and increases in cardiac index. The hemodynamic effects of nesiritide infusion were sustained over a duration of 1 week and the drug may be used without intensive monitoring in patients with decompensated HF. Treatment with dobutamine is indicated in patients in whom low cardiac output rather than elevated pulmonary pressure is the primary hemodynamic aberration. However, milrinone reduces left atrial congestion more effectively than dobutamine, and is well tolerated and effective when used in patients receiving beta-blockers. In-patient therapy for decompensated HF is a short term exercise for symptom relief and provides an opportunity to re-assess management in the continuum of care.
Collapse
Affiliation(s)
- R M Mills
- The Section of Clinical Cardiology and the Kaufman Center for Heart Failure, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA.
| | | |
Collapse
|
20
|
Porter JD, Merriam AP, Leahy P, Gong B, Khanna S. Dissection of temporal gene expression signatures of affected and spared muscle groups in dystrophin-deficient (mdx) mice. Hum Mol Genet 2003; 12:1813-21. [PMID: 12874102 DOI: 10.1093/hmg/ddg197] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Although dystrophin mutations are the proximate cause of Duchenne muscular dystrophy (DMD), interactions among heterogeneous downstream mechanisms may be key phenotypic determinants. Temporal gene expression profiling was used to identify and correlate diverse transcriptional patterns to one another and to the disease course, for both affected and spared muscle groups, in postnatal day 7-112 dystrophin-deficient (mdx) mice. While 719 transcripts were differentially expressed at one or more ages in leg muscle, only 56 genes were altered in the spared extraocular muscles (EOM). Contrasting molecular signatures of affected versus spared muscles provide compelling evidence that the absence of dystrophin alone is necessary but not sufficient to cause the patterned fibrosis, inflammation and failure of muscle regeneration characteristic of dystrophinopathy. Dystrophic and adaptive changes in the microarray profiles were further quantified using an aggregate disease load index (DLI) to measure stage-dependent transcriptional impact in both muscles. DLI analysis highlighted the divergent responses of EOM and leg muscle groups. Cellular process-specific DLIs in leg muscle identified positively correlated temporal expression profiles for some gene classes, and the independence of others, that are linked to major disease components. Data also showed a previously unrecognized transient and selective developmental delay in pre-necrotic mdx skeletal muscle that was confirmed by qPCR. Taken together, validation and targeting of signaling pathways responsible for the coordination of the fibrotic, proteolytic and inflammatory mechanisms shown here for mdx muscle may yield new therapeutic means of mitigating the devastating consequences of DMD.
Collapse
Affiliation(s)
- John D Porter
- Department of Ophthalmology, Case Western Reserve University and University Hospitals of Cleveland, 11100 Euclid Avenue, Cleveland, OH 44106, USA.
| | | | | | | | | |
Collapse
|
21
|
Faris RF, Henein MY, Coats AJS. Influence of gender and reported alcohol intake on mortality in nonischemic dilated cardiomyopathy. HEART DISEASE (HAGERSTOWN, MD.) 2003; 5:89-94. [PMID: 12713675 DOI: 10.1097/01.hdx.0000061702.79961.47] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Between 1994-1998, we retrospectively studied a cohort of 396 consecutive patients with dilated cardiomyopathy, mean age 53 +/- 15 years, 74% men. The history of alcohol intake was abstracted from the medical records. During a follow-up period of 42 +/- 26 months, 83 (76% men) patients died and 15 (80% men) underwent cardiac transplantation. Men were younger and more likely to have a history of excessive alcohol intake compared with women. Gender significantly modified the risk of alcohol consumption on mortality. Although alcohol intake increased the mortality risk in women, it was protective toward death in men (hazards ratios for death were 7.3 vs. 0.44). The effect of alcohol intake on outcome was reassessed after classifying the patients into 4 groups: group 1: life-long nondrinkers; group 2: former drinkers; group 3: moderate drinkers; and group 4: heavy drinkers. Similar findings were seen. This study demonstrated that the risk of reported alcohol intake on mortality is related to gender in patients with nonischemic heart failure. Our findings deserve further study, including a larger number of females, as a possible way to improve outcome in such patients.
Collapse
Affiliation(s)
- R F Faris
- Department of Clinical Cardiology, Royal Brompton Hospital, London, United Kingdom.
| | | | | |
Collapse
|
22
|
Ahmed A, Maisiak R, Allman RM, DeLong JF, Farmer R. Heart failure mortality among older Medicare beneficiaries: association with left ventricular function evaluation and angiotensin-converting enzyme inhibitor use. South Med J 2003; 96:124-9. [PMID: 12630634 DOI: 10.1097/01.smj.0000051271.11872.50] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Left ventricular function evaluation and angiotensin-converting enzyme (ACE) inhibitor use are the two basic indicators of heart failure quality of care. In this retrospective follow-up study, we analyzed the association between these two quality indicators and mortality in elderly hospitalized heart failure patients. METHODS The patients in our study were older Alabama Medicare beneficiaries discharged with a diagnosis of heart failure in 1994. Cox regression analyses, adjusted for various patient and care characteristics, were performed to estimate the overall mortality rate. RESULTS The mean age of the 1,090 patients in our study was 79+/-7.5 years. Both left ventricular function evaluation (hazard ratio, 0.83; 95% confidence interval, 0.705-0.976) and ACE inhibitor use (hazard ratio, 0.77; 95% confidence interval, 0.655-0.905) were associated with a lower 3-year mortality rate. Adjustment for various patient and care characteristics did not alter these associations. CONCLUSION Left ventricular function evaluation and ACE inhibitor use were each associated with increased survival time in older Medicare beneficiaries with heart failure.
Collapse
Affiliation(s)
- Ali Ahmed
- Division of Gerontology and Geriatric Medicine, Department of Medicine, Center for Aging, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-2041, USA.
| | | | | | | | | |
Collapse
|
23
|
Heyen JRR, Blasi ER, Nikula K, Rocha R, Daust HA, Frierdich G, Van Vleet JF, De Ciechi P, McMahon EG, Rudolph AE. Structural, functional, and molecular characterization of the SHHF model of heart failure. Am J Physiol Heart Circ Physiol 2002; 283:H1775-84. [PMID: 12384454 DOI: 10.1152/ajpheart.00305.2002] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heart failure is a complex multifactorial disease resulting in a myriad of progressive changes at the molecular, cellular, and physiological level. To better understand the mechanisms associated with the development of congestive heart failure, a comprehensive examination of the aging lean male spontaneously hypertensive, heart failure-prone rat (SHHF) was conducted. Myocardial function and structural integrity progressively diminished as evidenced by decreased ejection fraction and increased left ventricular volume measured using echocardiography. Functional and structural changes were accompanied by elevations in circulating inflammatory markers, including tumor necrosis factor-alpha (TNF-alpha), IL-6, and TNF receptors type 1 and 2. Increased systemic inflammatory marker levels were consistent with age-dependent changes in the expression pattern of genes that contribute to stress, inflammation, and the extracellular matrix in SHHF animals analyzed from age 4 to 18 mo. In summary, the SHHF rat shares many hallmark features of the human disease state and represents a key experimental model for the dissection of complex human heart failure pathophysiology.
Collapse
Affiliation(s)
- Jonathan R R Heyen
- Pharmacia Corporation, 800 N. Lindbergh Boulevard, St. Louis, MO 63167, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Yamamoto K, Masuyama T, Sakata Y, Nishikawa N, Mano T, Hori M. Prevention of diastolic heart failure by endothelin type A receptor antagonist through inhibition of ventricular structural remodeling in hypertensive heart. J Hypertens 2002; 20:753-61. [PMID: 11910313 DOI: 10.1097/00004872-200204000-00035] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Despite the clinical frequency of diastolic heart failure, its therapeutic strategy has not been established. Our recent study demonstrated activation of the endothelin (ET) system in a diastolic heart failure model with hypertension. Several studies have reported that ET type A (ETA) receptor antagonist improves systolic function and prevents systolic heart failure; however, its effects on diastolic heart failure are unknown. We investigated the effects of chronic administration of ET(A) receptor antagonist in diastolic heart failure. DESIGN AND METHODS Dahl-Iwai salt-sensitive rats fed on a high-salt diet from 7 weeks of age, in which congestive heart failure develops following hypertension without cardiac chamber dilatation or systolic dysfunction, were divided into groups with and without administration of a subdepressor dose of ET(A) receptor antagonist. RESULTS Hypertension induced compensatory left ventricular (LV) hypertrophy at 13 weeks in six untreated rats. Persistent pressure overload developed progressive LV hypertrophy and fibrosis from 13 to 19 weeks, resulting in elevated LV filling pressure and increased lung weight. Chronic ET(A) receptor blockade did not restrain compensatory LV hypertrophy at 13 weeks; however, it attenuated LV hypertrophy and fibrosis thereafter (n = 6). These beneficial effects resulted in the maintenance of normal LV filling pressure without changes in LV end-diastolic diameter, indicating prevention of LV stiffening. CONCLUSIONS Chronic ET(A) receptor blockade is likely to exert beneficial effects in diastolic failure through attenuation of the progression of LV hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Department of Internal Medicine and Therapeutics, Osaka University Graduate School of Medicine, Suita, Japan.
| | | | | | | | | | | |
Collapse
|
25
|
Stawowy P, Blaschke F, Pfautsch P, Goetze S, Lippek F, Wollert-Wulf B, Fleck E, Graf K. Increased myocardial expression of osteopontin in patients with advanced heart failure. Eur J Heart Fail 2002; 4:139-46. [PMID: 11959041 DOI: 10.1016/s1388-9842(01)00237-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The expression of the adhesion protein osteopontin (OP) is associated with cardiac hypertrophy and is significantly increased after transition to heart failure in experimental animal models. We, therefore, hypothesized that OP could be upregulated in heart failure in humans. In the present study, we investigated the expression of OP in myocardial biopsies obtained from patients with heart failure due to dilated cardiomyopathy (mean LVEF=30.3+/-4.4%, mean+/-S.D., n=10, group A) compared to patients with a normal left-ventricular ejection fraction (mean LVEF=61+/-11.2%, n=9; group B). Myocardial immunoreactivity for OP was examined using two different antibodies against OP. The expression of cardiac myocyte OP was significantly upregulated in group A in comparison to group B (P<0.0001). Both groups also displayed OP immunoreactivity in non-myocytes, including vascular smooth muscle cells and cardiac fibroblasts (P=not significant). Statistical analysis revealed a significant correlation of increased OP immunoreactivity in cardiac myocytes of patients with impaired left ventricular function, assessed by hemodynamic data (LVEF, RVEF, LVESVI, LVEDVI and LVEDP, R=-0.828, -0.671, 0.751, 0.685 and 0.461, respectively; all P<0.05). Furthermore, OP expression correlated with cardiac myocyte hypertrophy (mean diameter 21.0+/-1.8 microm in group A and 16.6+/-2.1 microm in group B; P<0.0001). In conclusion, the present study indicates, that factors and/or mechanisms involved in heart failure in patients with dilated cardiomyopathy, lead to induction of OP expression in humans.
Collapse
Affiliation(s)
- Philipp Stawowy
- Department of Medicine/Cardiology, German Heart Institute Berlin, Augustenburger Platz 1, D-13353, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Mavroidis M, Capetanaki Y. Extensive induction of important mediators of fibrosis and dystrophic calcification in desmin-deficient cardiomyopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 160:943-52. [PMID: 11891192 PMCID: PMC1867168 DOI: 10.1016/s0002-9440(10)64916-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Mice lacking the intermediate filament protein desmin demonstrate abnormal mitochondria behavior, disruption of muscle architecture, and myocardial degeneration with extensive calcium deposits and fibrosis. These abnormalities are associated with cardiomyocyte hypertrophy, cardiac chamber dilation and eventually with heart failure. In an effort to elucidate the molecular mechanisms leading to the observed pathogenesis, we have analyzed gene expression changes in cardiac tissue using differential display polymerase chain reaction and cDNA atlas array methods. The most substantial changes were found in genes coding the small extracellular matrix proteins osteopontin and decorin that are dramatically induced in the desmin-null myocardium. We further analyzed their expression pattern both at the RNA and protein levels and we compared their spatial expression with the onset of calcification. Extensive osteopontin localization is observed by immunohistochemistry in the desmin-null myocardium in areas with massive myocyte death, as well as in hypercellular regions with variable degrees of calcification and fibrosis. Osteopontin is consistently co-localized with calcified deposits, which progressively are transformed to psammoma bodies surrounded by decorin, especially in the right ventricle. These data together with the observed up-regulation of transforming growth factor-beta1 and angiotensin-converting enzyme, could explain the extensive fibrosis and dystrophic calcification observed in the heart of desmin-null mice, potentially crucial events leading to heart failure.
Collapse
Affiliation(s)
- Manolis Mavroidis
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
27
|
Piller LB, Davis BR, Cutler JA, Cushman WC, Wright JT, Williamson JD, Leenen FHH, Einhorn PT, Randall OS, Golden JS, Haywood LJ. Validation of Heart Failure Events in the Antihypertensive and Lipid Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) Participants Assigned to Doxazosin and Chlorthalidone. CURRENT CONTROLLED TRIALS IN CARDIOVASCULAR MEDICINE 2002; 3:10. [PMID: 12459039 PMCID: PMC149403 DOI: 10.1186/1468-6708-3-10] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2002] [Accepted: 11/14/2002] [Indexed: 11/10/2022]
Abstract
BACKGROUND: The Antihypertensive and Lipid Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) is a randomized, double-blind, active-controlled trial designed to compare the rate of coronary heart disease events in high-risk hypertensive participants initially randomized to a diuretic (chlorthalidone) versus each of three alternative antihypertensive drugs: alpha-adrenergic blocker (doxazosin), ACE-inhibitor (lisinopril), and calcium-channel blocker (amlodipine). Combined cardiovascular disease risk was significantly increased in the doxazosin arm compared to the chlorthalidone arm (RR 1.25; 95% CI, 1.17-1.33; P <.001), with a doubling of heart failure (fatal, hospitalized, or non-hospitalized but treated) (RR 2.04; 95% CI, 1.79-2.32; P <.001). Questions about heart failure diagnostic criteria led to steps to validate these events further. METHODS AND RESULTS: Baseline characteristics (age, race, sex, blood pressure) did not differ significantly between treatment groups (P <.05) for participants with heart failure events. Post-event pharmacologic management was similar in both groups and generally conformed to accepted heart failure therapy. Central review of a small sample of cases showed high adherence to ALLHAT heart failure criteria. Of 105 participants with quantitative ejection fraction measurements provided, (67% by echocardiogram, 31% by catheterization), 29/46 (63%) from the chlorthalidone group and 41/59 (70%) from the doxazosin group were at or below 40%. Two-year heart failure case-fatalities (22% and 19% in the doxazosin and chlorthalidone groups, respectively) were as expected and did not differ significantly (RR 0.96; 95% CI, 0.67-1.38; P = 0.83). CONCLUSION: Results of the validation process supported findings of increased heart failure in the ALLHAT doxazosin treatment arm compared to the chlorthalidone treatment arm.
Collapse
Affiliation(s)
- Linda B Piller
- The University of Texas School of Public Health, Houston, TX, USA
| | - Barry R Davis
- The University of Texas School of Public Health, Houston, TX, USA
| | | | | | - Jackson T Wright
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | | | - Paula T Einhorn
- National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | | | - John S Golden
- Kaiser Permanente, Mid-Atlantic States, Washington, DC, USA
| | - L Julian Haywood
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
28
|
Caldwell MA, Dracup K. Team management of heart failure: the emerging role of exercise, and implications for cardiac rehabilitation centers. JOURNAL OF CARDIOPULMONARY REHABILITATION 2001; 21:273-9. [PMID: 11591041 DOI: 10.1097/00008483-200109000-00004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- M A Caldwell
- University of California San Francisco, PO Box 1545, Healdsburg, CA 95448, USA.
| | | |
Collapse
|
29
|
Chiba S, Rashid MM, Okamoto H, Shiraiwa H, Kon S, Maeda M, Murakami M, Inobe M, Kitabatake A, Chambers AF, Uede T. The role of osteopontin in the development of granulomatous lesions in lung. Microbiol Immunol 2000; 44:319-32. [PMID: 10832978 DOI: 10.1111/j.1348-0421.2000.tb02501.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Osteopontin (OPN) has been shown to be expressed by cells in granulomas of various origins, but whether it plays a functional role in granuloma formation is not known. Here we used a cardiomyopathic hamster (TO2) model, to test the hypothesis that OPN contributes functionally to granuloma development. We immunized cardiomyopathic and normal hamsters by subcutaneous injection of bovine serum albumin in complete Freund's adjuvant, and assessed various tissues for both OPN RNA expression and granuloma formation. Cardiomyopathic hamsters expressed OPN, and formed granulomatous lesions, in heart tissue in both immunized and untreated animals. In addition, immunization induced expression of OPN in lung and lymph nodes of cardiomyopathic (but not normal) hamsters, and also induced granuloma formation in these organs. To test whether OPN expression could play a functional role in inducing granulomas, we produced an adenoviral vector containing the murine OPN gene, and introduced this vector intratracheally into the lungs of normal hamsters. The OPN-containing vector, but not the control vector, induced pulmonary granuloma formation. These studies provided direct in vivo evidence that OPN can contribute functionally to the formation of granulomatous lesions, and suggest that OPN expression may be a common factor involved in formation of granulomas of various origin.
Collapse
Affiliation(s)
- S Chiba
- Section of Immunopathogenesis, Institute of Immunological Science, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Denovan-Wright EM, Ferrier GR, Robertson HA, Howlett SE. Increased expression of the gene for alpha-interferon-inducible protein in cardiomyopathic hamster heart. Biochem Biophys Res Commun 2000; 267:103-8. [PMID: 10623582 DOI: 10.1006/bbrc.1999.1938] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiomyopathic (CM) hamsters have a disruption in the delta-sarcoglycan gene which leads to progressive cardiac necrosis by 30 to 40 days of age, hypertrophy by 120 days, and heart failure by 250 days. We used differential display to detect other changes in mRNA levels in 30-, 60-, and 90-day-old wild-type and CM hamsters. We identified a 400-bp cDNA with sequence similarity to the human alpha-interferon-inducible protein (p27). This cDNA annealed with a 570-base mRNA whose steady-state levels were increased in 30-, 60-, and 90-day-old CM compared to wild-type heart. Increased expression of this hamster homolog of p27 (p27-h) was detected in CM hamster cardiac and skeletal muscle at 60 days of age but not in liver, kidney, or brain. Thus, an inherited defect in CM hamsters leads to increased expression of p27-h in advance of the development of hypertrophy and heart failure.
Collapse
Affiliation(s)
- E M Denovan-Wright
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, B3H 4H7, Canada
| | | | | | | |
Collapse
|
31
|
Doi R, Masuyama T, Yamamoto K, Doi Y, Mano T, Sakata Y, Ono K, Kuzuya T, Hirota S, Koyama T, Miwa T, Hori M. Development of different phenotypes of hypertensive heart failure: systolic versus diastolic failure in Dahl salt-sensitive rats. J Hypertens 2000; 18:111-20. [PMID: 10678551 DOI: 10.1097/00004872-200018010-00016] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE There are two phenotypes of heart failure, systolic failure and isolated diastolic heart failure with preserved left ventricular systolic function. Although isolated diastolic heart failure frequently occurs, there are only models for diastolic dysfunction unassociated with heart failure and models with overt diastolic heart failure have not been established. We attempted to develop two different models, i.e. diastolic and systolic failure models, based on hypertension. MATERIALS AND METHODS Dahl salt-sensitive rats were placed on 8% NaCl diet from 7 weeks old (7-week starting group) or 8 weeks old (8-week starting group). As an age-matched control, Dahl salt-sensitive rats were consistently placed on normal chow. In these rats, echocardiogram was serially recorded, followed by hemodynamic and histological studies. RESULTS The 7-week starting rats showed a steep elevation in blood pressure and progressive left ventricular hypertrophy, and fell into overt heart failure at approximately 19 weeks. The development of heart failure was not associated with a decrease in left ventricular midwall fractional shortening or an increase in left ventricular end-diastolic dimension as compared with the age-matched control, which mimics the characteristics of clinically observed isolated diastolic heart failure. The 8-week starting rats showed a gradual rise in blood pressure and less progressive left ventricular hypertrophy, and fell into heart failure at approximately 26 weeks with a decrease in mid-wall fractional shortening and an increase in left ventricular end-diastolic dimension. Hemodynamic and histological studies at failing stage revealed comparable elevation of left ventricular end-diastolic pressure and comparable left ventricular fibrosis in both groups. CONCLUSION These two different models of overt heart failure may be useful as models of isolated diastolic heart failure and systolic heart failure based on the same hypertensive heart disease, respectively, and may contribute to discrimination of the mechanisms of the development of the two different phenotypes of heart failure.
Collapse
Affiliation(s)
- R Doi
- Department of Internal Medicine and Therapeutics, Osaka University Graduate School of Medicine, Suita, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Zhao G, Zhang X, Smith CJ, Xu X, Ochoa M, Greenhouse D, Vogel T, Curran C, Hintze TH. Reduced coronary NO production in conscious dogs after the development of alloxan-induced diabetes. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:H268-78. [PMID: 10409206 DOI: 10.1152/ajpheart.1999.277.1.h268] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of nitric oxide (NO) in the control of coronary blood flow (CBF) during the development of diabetes is unknown. To study this, mongrel dogs were chronically instrumented using sterile techniques for measurements of systemic hemodynamics and CBF. With heart rate controlled (150 beats/min), veratrine (1-10 micrograms/kg) caused dose-dependent increases in CBF; e.g., 5 mirograms/kg of veratrine increased CBF by 57 +/- 7% from 41 +/- 1.3 ml/min (P < 0.05). The dogs developed diabetes 4-5 wk after injection of alloxan (40-60 mg/kg iv, blood glucose levels were 384 +/- 18 mg/dl). After diabetes the same doses of veratrine caused smaller increases in CBF; i.e., 5 micrograms/kg of veratrine increased CBF by 32 +/- 2% (P < 0.05 compared with control) from 28 +/- 4 ml/min. ACh- and adenosine-induced coronary vasodilation were reduced after diabetes as well. In anesthetized dogs after diabetes, vagal stimulation caused smaller increases in CBF. ACh and bradykinin caused smaller increases in NO(-)(2) production in coronary microvessels from diabetic dogs. Furthermore, despite the fact that mRNA for endothelial cell NO synthase from the aorta was increased twofold with the use of Northern blotting, the protein for aortic endothelial constitutive NO synthase was reduced by 66% after diabetes, as determined by Western blotting. Our results indicate that the NO-dependent coronary vasodilation by the Bezold-Jarisch reflex is impaired in conscious dogs after diabetes. The mechanism responsible for the impaired endothelium-dependent coronary vasodilation is most likely the decreased release of NO from the endothelium.
Collapse
Affiliation(s)
- G Zhao
- Department of Physiology, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
In this review we consider available information on the cardio- and cytoprotective properties and mechanism of action of trimetazidine, an antianginal drug. Two points in particular are addressed: the advantages of trimetazidine over classic drugs and its possible use for the long-term treatment of hypertrophic cardiomyopathy, described as an ischemic disorder.
Collapse
Affiliation(s)
- N D'hahan
- Département de Biologie Cellulaire et Moléculaire, CEA-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
34
|
Davison G, Whittaker P, Wickline SA. Captopril Therapy Limits Ventricular Remodeling But Does Not Alter Myocardial Collagen Fiber Morphology of Cardiomyopathic Hamsters. Cardiovasc Pathol 1997; 6:307-13. [DOI: 10.1016/s1054-8807(97)00003-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
35
|
Stanley M. Current trends in the clinical management of an old enemy: congestive heart failure in the elderly. AACN CLINICAL ISSUES 1997; 8:616-26; quiz 649-51. [PMID: 9392718 DOI: 10.1097/00044067-199711000-00013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Much has been learned in the past decade, through quality medical and nursing research, regarding the pathophysiology and management of congestive heart failure. Primary care practitioners are challenged to apply the latest data to the clinical management of these patients when there is clear evidence of improvement in the control of symptoms and quality of life. Because of the complexities of the disease process, older adults with congestive heart failure require a comprehensive approach that is particularly well suited to the knowledge and skills of the advanced practice nurse.
Collapse
Affiliation(s)
- M Stanley
- Southern Illinois University at Edwardsville, USA
| |
Collapse
|
36
|
Uede T, Katagiri Y, Iizuka J, Murakami M. Osteopontin, a coordinator of host defense system: a cytokine or an extracellular adhesive protein? Microbiol Immunol 1997; 41:641-8. [PMID: 9343813 DOI: 10.1111/j.1348-0421.1997.tb01906.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- T Uede
- Section of Immunopathogenesis, Hokkaido University, Sapporo, Japan
| | | | | | | |
Collapse
|
37
|
Ashizawa N, Graf K, Do YS, Nunohiro T, Giachelli CM, Meehan WP, Tuan TL, Hsueh WA. Osteopontin is produced by rat cardiac fibroblasts and mediates A(II)-induced DNA synthesis and collagen gel contraction. J Clin Invest 1996; 98:2218-27. [PMID: 8941637 PMCID: PMC507670 DOI: 10.1172/jci119031] [Citation(s) in RCA: 188] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Angiotensin II (AII) is a critical factor in cardiac remodeling which involves hypertrophy, fibroblast proliferation, and extracellular matrix production. However, little is known about the mechanism by which AII accelerates these responses. Osteopontin is an acidic phosphoprotein with RGD (arginine-glycine-aspartate) sequences that are involved in the vascular smooth muscle cell remodeling process. We identified the presence of osteopontin mRNA and protein in cultured rat cardiac fibroblasts and its prominent regulation by AII (10(-11) M). Osteopontin message levels were increased fourfold (P < 0.01) and protein fivefold (P < 0.05) at 24 h after addition of AII (10(-7) M). This response was inhibited by the AT1 receptor blocker, losartan. Osteopontin mRNA levels were increased in hypertrophied ventricles from animals with renovascular hypertension (1.6-fold, P < 0.05) and aortic banding (2.9-fold, P < 0.05). To examine the function of osteopontin, we determined its effects on (a) the ability of cardiac fibroblasts to contract three-dimensional collagen gels and (b) cardiac fibroblast growth. A monoclonal antibody against osteopontin partially blocked AII-induced three-dimensional collagen gel contraction by cardiac fibroblasts (64+/-4 vs. 86+/-5% in the presence of antibody, P < 0.05), while osteopontin itself promoted contraction of the gels by fibroblasts (71+/-5%, P < 0.05 compared with control). Either a monoclonal antibody against beta3 integrin which is a ligand for osteopontin or the RGD peptide blocked both AII and osteopontin-induced collagen gel contraction. Thus, the osteopontin RGD sequence binds to beta3 integrins on the fibroblast to promote fibroblast binding to collagen. All induced a threefold increase in DNA synthesis of cardiac fibroblasts, which was completely blocked by antibodies against osteopontin and beta3 integrin, or by RGD peptide, but not by controls. Thus, All-induced growth of cardiac fibroblasts also requires osteopontin engagement of the beta3 integrin. Taken together, these results provide the first evidence that osteopontin is a potentially important mediator of AII regulation of cardiac fibroblast behavior in the cardiac remodeling process.
Collapse
Affiliation(s)
- N Ashizawa
- University of Southern California School of Medicine, Department of Medicine, Los Angeles 90033, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Singh K, Balligand JL, Fischer TA, Smith TW, Kelly RA. Glucocorticoids increase osteopontin expression in cardiac myocytes and microvascular endothelial cells. Role in regulation of inducible nitric oxide synthase. J Biol Chem 1995; 270:28471-8. [PMID: 7499354 DOI: 10.1074/jbc.270.47.28471] [Citation(s) in RCA: 110] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
In heart muscle, the cytokine-inducible isoform of nitric oxide synthase (NOS2) is expressed in both cardiac myocytes and microvascular endothelial cells (CMEC). mRNA levels for both NOS2 and for osteopontin, a multifunctional extracellular matrix phosphoprotein containing and RGD integrin binding domain, are increased in cardiac muscle following intraperitoneal injection of adult rats with lipopolysaccharide. In vitro, interleukin-1 beta and interferon-gamma increased osteopontin mRNA levels in CMEC as well as NOS2 expression in both CMEC and cardiac myocytes. However, osteopontin mRNA levels in heart muscle in vivo, and in cardiac myocytes and CMEC in vitro, also are increased 10-30-fold by the synthetic glucocorticoid dexamethasone, an agent that suppresses cytokine induction of NOS2 in both cell types. The hexapeptide GRGDSP, which interrupts binding of RGD-containing proteins to cell surface integrins, increased NOS2 mRNA, while a synthetic osteopontin peptide analogue decreased NOS2 mRNA and protein levels in both cytokine-pretreated cardiac myocytes and CMEC cultures. Also, transfection with a full-length antisense-osteopontin cDNA in cytokine-pretreated CMEC decreased endogenous osteopontin mRNA and increased NOS2 mRNA levels. These results suggest that osteopontin could regulate the location and extent of NOS2 induction in the heart. Increased expression of osteopontin also may be one mechanism by which glucocorticoids suppress NOS2 activity in cardiac myocytes and microvascular endothelial cells.
Collapse
Affiliation(s)
- K Singh
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|